1
|
Thomson R, Close K, Riley A, Batstone DJ, Oehmen A. Metabolic modelling of anaerobic amino acid uptake and storage by fermentative polyphosphate accumulating organisms. WATER RESEARCH 2025; 280:123512. [PMID: 40138861 DOI: 10.1016/j.watres.2025.123512] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 12/13/2024] [Revised: 03/03/2025] [Accepted: 03/17/2025] [Indexed: 03/29/2025]
Abstract
Existing enhanced biological phosphorus removal (EBPR) models do not fully describe the metabolism of fermentative polyphosphate accumulating organisms (fPAOs), particularly under mixed substrate conditions representative of fermentation-enhanced EBPR (F-EBPR) processes. This study presents a steady-state metabolic model integrating anaerobic amino acid (AA) fermentation and storage processes in fPAOs. The model identifies key metabolic interactions underlying fPAO metabolism, prioritising substrate accumulation over fermentation. This results in significant changes to ATP and reduction-oxidation (redox) flows as compared to when relying on previous AA fermentation models typically used to describe fPAO metabolism, with medium-chain-length (MCL) polyhydroxyalkanoate (PHA) formation and polyphosphate (polyP) consumption acting as important electron and energy management mechanisms, respectively. Succinate, rather than volatile fatty acids (VFAs), was identified as the more likely synergetic substrate between fermentative and conventional PAOs (cPAOs) under these conditions. Moreover, conditions favourable of VFA efflux by fPAOs may also favour a shift away from a polyP accumulation to a fermentation dominant metabolism. Further work is required to verify the role of MCL-PHA fractions, alongside the contribution of free intracellular AA accumulation as compared to polymers such as cyanophycin or polyglutamate on fPAO metabolism. This metabolic model provides a framework for better understanding the role of fPAOs and their interactions with cPAOs within EBPR processes, informing future modelling and optimisation of F-EBPR systems.
Collapse
Affiliation(s)
- R Thomson
- School of Chemical Engineering, Australian Centre for Water and Environmental Biotechnology, University of Queensland, St Lucia, QLD 4072, Australia
| | - K Close
- School of Chemical Engineering, Australian Centre for Water and Environmental Biotechnology, University of Queensland, St Lucia, QLD 4072, Australia
| | - A Riley
- School of Chemical Engineering, Australian Centre for Water and Environmental Biotechnology, University of Queensland, St Lucia, QLD 4072, Australia
| | - D J Batstone
- Australian Centre for Water and Environmental Biotechnology, University of Queensland, St Lucia, QLD 4072, Australia
| | - A Oehmen
- School of Chemical Engineering, Australian Centre for Water and Environmental Biotechnology, University of Queensland, St Lucia, QLD 4072, Australia.
| |
Collapse
|
2
|
Thomson R, Le C, Wang L, Batstone DJ, Zhou Y, Oehmen A. Higher order volatile fatty acid metabolism and atypical polyhydroxyalkanoate production in fermentation-enhanced biological phosphorus removal. WATER RESEARCH 2025; 280:123503. [PMID: 40121909 DOI: 10.1016/j.watres.2025.123503] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 05/03/2024] [Revised: 03/11/2025] [Accepted: 03/13/2025] [Indexed: 03/25/2025]
Abstract
Enhanced biological phosphorus removal (EBPR) is an established wastewater treatment process, but its wider implementation has been limited by factors like high temperature and low carbon availability. Fermentation-enhanced EBPR (F-EBPR) processes have shown promise in addressing these limitations, but the underlying mechanisms are not fully understood. This study investigates the metabolism of higher order (C4-5) volatile fatty acids (VFAs) in F-EBPR systems using a combination of carbon isotope labelling and shotgun metagenomic sequencing analyses. Results show that butyrate (HBu) uptake leads to the formation of both typical (C4-5) and atypical (C6+) polyhydroxyalkanoates (PHAs) through a combination ofβ-oxidation and standard condensation pathways, while the putative role of HBu oxidisers were identified relative to substrate composition in F-EBPR processes. Metagenomic analysis reveals the presence of genes required for higher order VFA metabolism in both polyphosphate accumulating organisms (PAOs) and glycogen accumulating organisms (GAOs). This study also highlights the limitations of current models in describing F-EBPR processes and emphasises the need for improved models that account for higher order VFA metabolism and microbial community dynamics.
Collapse
Affiliation(s)
- R Thomson
- School of Chemical Engineering, University of Queensland, St Lucia QLD 4072, Australia
| | - C Le
- Asian School of the Environment, Nanyang Technological University, 637141, Singapore
| | - L Wang
- College of Chemistry and Chemical Engineering, Shanghai University of Engineering Science, Shanghai 201600, PR China
| | - D J Batstone
- Australian Centre for Water and Environmental Biotechnology, University of Queensland, St Lucia QLD 4072, Australia
| | - Y Zhou
- Advanced Environmental Biotechnology Center, Nanyang Technological University, 637141, Singapore.
| | - A Oehmen
- School of Chemical Engineering, University of Queensland, St Lucia QLD 4072, Australia.
| |
Collapse
|
3
|
Singhal R, Izquierdo P, Ranaweera T, Segura Abá K, Brown BN, Lehti-Shiu MD, Shiu SH. Using supervised machine-learning approaches to understand abiotic stress tolerance and design resilient crops. Philos Trans R Soc Lond B Biol Sci 2025; 380:20240252. [PMID: 40439305 PMCID: PMC12121380 DOI: 10.1098/rstb.2024.0252] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 01/14/2025] [Accepted: 01/15/2025] [Indexed: 06/02/2025] Open
Abstract
Abiotic stresses such as drought, heat, cold, salinity and flooding significantly impact plant growth, development and productivity. As the planet has warmed, these abiotic stresses have increased in frequency and intensity, affecting the global food supply and making it imperative to develop stress-resilient crops. In the past 20 years, the development of omics technologies has contributed to the growth of datasets for plants grown under a wide range of abiotic environments. Integration of these rapidly growing data using machine-learning (ML) approaches can complement existing breeding efforts by providing insights into the mechanisms underlying plant responses to stressful conditions, which can be used to guide the design of resilient crops. In this review, we introduce ML approaches and provide examples of how researchers use these approaches to predict molecular activities, gene functions and genotype responses under stressful conditions. Finally, we consider the potential and challenges of using such approaches to enable the design of crops that are better suited to a changing environment.This article is part of the theme issue 'Crops under stress: can we mitigate the impacts of climate change on agriculture and launch the 'Resilience Revolution'?'.
Collapse
Affiliation(s)
- Rajneesh Singhal
- Department of Plant Biology, Michigan State University, East Lansing, MI48824, USA
| | - Paulo Izquierdo
- Department of Plant Biology, Michigan State University, East Lansing, MI48824, USA
- DOE Great Lakes Bioenergy Research Center, Michigan State University, East Lansing, MI48824, USA
| | - Thilanka Ranaweera
- Department of Plant Biology, Michigan State University, East Lansing, MI48824, USA
- DOE Great Lakes Bioenergy Research Center, Michigan State University, East Lansing, MI48824, USA
| | - Kenia Segura Abá
- DOE Great Lakes Bioenergy Research Center, Michigan State University, East Lansing, MI48824, USA
- Genetics and Genome Sciences Program, Michigan State University, East Lansing, MI48824, USA
| | - Brianna N.I. Brown
- Department of Plant Biology, Michigan State University, East Lansing, MI48824, USA
| | | | - Shin-Han Shiu
- Department of Plant Biology, Michigan State University, East Lansing, MI48824, USA
- DOE Great Lakes Bioenergy Research Center, Michigan State University, East Lansing, MI48824, USA
- Genetics and Genome Sciences Program, Michigan State University, East Lansing, MI48824, USA
- Department of Computational Mathematics, Science, and Engineering, Michigan State University, East Lansing, MI48824, USA
| |
Collapse
|
4
|
Naidoo TJ, Senzani S, Singh R, Pillay B, Pillay M. Mycobacterium tuberculosis curli pili (MTP) and heparin-binding hemagglutinin adhesin (HBHA) facilitate regulation of central carbon metabolism, enhancement of ATP synthesis and cell wall biosynthesis. Arch Microbiol 2025; 207:156. [PMID: 40437078 PMCID: PMC12119724 DOI: 10.1007/s00203-025-04352-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/18/2025] [Revised: 04/14/2025] [Accepted: 04/30/2025] [Indexed: 06/01/2025]
Abstract
Functional 'omics' studies previously identified the M. tuberculosis surface located adhesins, heparin-binding hemagglutinin adhesin (HBHA) and curli pili (MTP) as significant potential targets for the design of tuberculosis (TB) point-of-care diagnostics, effective drugs, and vaccines. Little is known on the effect of these adhesins on the pathogen's transcriptome. The current study, via transcriptomics, elucidated whether the deletion of the single genes, hbhA and mtp, and double genes, hbhA-mtp, via specialised transduction, affected global bacterial gene expression. RNA sequencing of M. tuberculosis wild-type V9124 (WT), single and double deletion HBHA and MTP mutant strains were confirmed by reverse transcription quantitative polymerase chain reaction (RT-qPCR) on selected genes, and a functional bacterial ATP bioluminescence assay. The 43 significantly differentially expressed genes amongst the deletion mutants were functionally categorized into central carbon metabolism (CCM), cell wall biosynthesis and cell wall transport and processes. The increased expression of genes associated with ATP synthase and cell wall processes were confirmed by RT-qPCR. In the absence of the adhesins, a decreased ATP concentration was observed suggesting either increased utilization or alterations to the proton motive force (PMF) that resulted in a potential inhibition of ATP synthesis. Therefore, deletions of the mtp and hbhA genes were associated with significant perturbations in CCM regulation/function, and transport of proteins to the cell wall, indicating the significant contribution of these adhesins in fundamental processes contributing to TB pathogenesis. Thus, this study indicates that MTP and HBHA influence gene expression in M. tuberculosis and represent important targets for TB diagnostic/therapeutic interventions and should be investigated as vaccine and adjunctive therapies.
Collapse
Affiliation(s)
- T J Naidoo
- Medical Microbiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu- Natal, Doris Duke Medical Research Institute, 1st Floor, Congella, Private Bag 7, Durban, 4013, South Africa
| | - S Senzani
- Medical Microbiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu- Natal, Doris Duke Medical Research Institute, 1st Floor, Congella, Private Bag 7, Durban, 4013, South Africa
| | - R Singh
- Department of Medical Microbiology, National Health Laboratory Service, College of Health Sciences, University of KwaZulu- Natal, 1st Floor, Congella, Private Bag 7, George Campbell BuildingDurban, 4013, South Africa
| | - B Pillay
- Microbiology, School of Life Sciences, College of Agriculture, Engineering and Science, University of KwaZulu- Natal, Westville Campus, Private Bag X54001, Durban, 4000, South Africa
| | - M Pillay
- Medical Microbiology, School of Laboratory Medicine and Medical Sciences, College of Health Sciences, University of KwaZulu- Natal, Doris Duke Medical Research Institute, 1st Floor, Congella, Private Bag 7, Durban, 4013, South Africa.
| |
Collapse
|
5
|
Hajnajafi K, Iqbal MA. Mass-spectrometry based metabolomics: an overview of workflows, strategies, data analysis and applications. Proteome Sci 2025; 23:5. [PMID: 40420110 PMCID: PMC12105183 DOI: 10.1186/s12953-025-00241-8] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/05/2024] [Accepted: 03/26/2025] [Indexed: 05/28/2025] Open
Abstract
BACKGROUND Metabolomics, a burgeoning field within systems biology, focuses on the comprehensive study of small molecules present in biological systems. Mass spectrometry (MS) has emerged as a powerful tool for metabolomic analysis due to its high sensitivity, resolution, and ability to characterize a wide range of metabolites thus offering deep insights into the metabolic profiles of living systems. AIM OF REVIEW This review provides an overview of the methodologies, workflows, strategies, data analysis techniques, and applications associated with mass spectrometry-based metabolomics. KEY SCIENTIFIC CONCEPTS OF REVIEW We discuss workflows, key strategies, experimental procedures, data analysis techniques, and diverse applications of metabolomics in various research domains. Nuances of sample preparation, metabolite extraction, separation using chromatographic techniques, mass spectrometry analysis, and data processing are elaborated. Moreover, standards, quality controls, metabolite annotation, software for statistical and pathway analysis are also covered. In conclusion, this review aims to facilitate the understanding and adoption of mass spectrometry-based metabolomics by newcomers and researchers alike by providing a foundational understanding and insights into the current state and future directions of this dynamic field.
Collapse
Affiliation(s)
- Kosar Hajnajafi
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates
| | - Mohammad Askandar Iqbal
- Thumbay Research Institute for Precision Medicine, Gulf Medical University, Ajman, United Arab Emirates.
- College of Medicine, Gulf Medical University, Ajman, United Arab Emirates.
| |
Collapse
|
6
|
Tibocha-Bonilla JD, Gandhi V, Lieng C, Moyne O, Santibáñez-Palominos R, Zengler K. Model of metabolism and gene expression predicts proteome allocation in Pseudomonas putida. NPJ Syst Biol Appl 2025; 11:55. [PMID: 40413180 PMCID: PMC12103522 DOI: 10.1038/s41540-025-00521-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2024] [Accepted: 04/20/2025] [Indexed: 05/27/2025] Open
Abstract
The genome-scale model of metabolism and gene expression (ME-model) for Pseudomonas putida KT2440, iPpu1676-ME, provides a comprehensive representation of biosynthetic costs and proteome allocation. Compared to a metabolic-only model, iPpu1676-ME significantly expands on gene expression, macromolecular assembly, and cofactor utilization, enabling accurate growth predictions without additional constraints. Multi-omics analysis using RNA sequencing and ribosomal profiling data revealed translational prioritization in P. putida, with core pathways, such as nicotinamide biosynthesis and queuosine metabolism, exhibiting higher translational efficiency, while secondary pathways displayed lower priority. Notably, the ME-model significantly outperformed the M-model in alignment with multi-omics data, thereby validating its predictive capacity. Thus, iPpu1676-ME offers valuable insights into P. putida's proteome allocation and presents a powerful tool for understanding resource allocation in this industrially relevant microorganism.
Collapse
Affiliation(s)
- Juan D Tibocha-Bonilla
- Bioinformatics and Systems Biology Graduate Program, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0760, USA
| | - Vishant Gandhi
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093-0412, USA
| | - Chloe Lieng
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0760, USA
| | - Oriane Moyne
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0760, USA
| | | | - Karsten Zengler
- Department of Bioengineering, University of California, San Diego, La Jolla, CA, 92093-0412, USA.
- Department of Pediatrics, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0760, USA.
- Center for Microbiome Innovation, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0403, USA.
- Program in Materials Science and Engineering, University of California, San Diego, 9500 Gilman Drive, La Jolla, CA, 92093-0418, USA.
| |
Collapse
|
7
|
Leonidou N, Renz A, Winnerling B, Grekova A, Grein F, Dräger A. Genome-scale metabolic model of Staphylococcus epidermidis ATCC 12228 matches in vitro conditions. mSystems 2025:e0041825. [PMID: 40396730 DOI: 10.1128/msystems.00418-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2025] [Accepted: 04/15/2025] [Indexed: 05/22/2025] Open
Abstract
Staphylococcus epidermidis, a commensal bacterium inhabiting collagen-rich areas like human skin, has gained significance due to its probiotic potential in the nasal microbiome and as a leading cause of nosocomial infections. While infrequently leading to severe illnesses, S. epidermidis exerts a significant influence, particularly in its close association with implant-related infections and its role as a classic opportunistic biofilm former. Understanding its opportunistic nature is crucial for developing novel therapeutic strategies, addressing both its beneficial and pathogenic aspects, and alleviating the burdens it imposes on patients and healthcare systems. Here, we employ genome-scale metabolic modeling as a powerful tool to elucidate the metabolic capabilities of S. epidermidis. We created a comprehensive computational resource for understanding the organism's growth conditions within diverse habitats by reconstructing and analyzing a manually curated and experimentally validated metabolic model. The final network, iSep23, incorporates 1,415 reactions, 1,051 metabolites, and 705 genes, adhering to established community standards and modeling guidelines. Benchmarking with the Metabolic Model Testing suite yields a high score, indicating the model's remarkable semantic quality. Following the findable, accessible, interoperable, and reusable (FAIR) data principles, iSep23 becomes a valuable and publicly accessible asset for subsequent studies. Growth simulations and carbon source utilization predictions align with experimental results, showcasing the model's predictive power. Ultimately, this work provides a robust foundation for future research aimed at both exploiting the probiotic potential and mitigating the pathogenic risks posed by S. epidermidis. IMPORTANCE Staphylococcus epidermidis, a bacterium commonly found on human skin, has shown probiotic effects in the nasal microbiome and is a notable causative agent of hospital-acquired infections. While these infections are typically non-life-threatening, their economic impact is considerable, with annual costs reaching billions of dollars in the United States. To better understand its opportunistic nature, we employed genome-scale metabolic modeling to construct a detailed network of S. epidermidis's metabolic capabilities. This model, comprising over a thousand reactions, metabolites, and genes, adheres to established standards and demonstrates solid benchmarking performance. Following the findable, accessible, interoperable, and reusable (FAIR) data principles, the model provides a valuable resource for future research. Growth simulations and predictions closely match experimental data, underscoring the model's predictive accuracy. Overall, this work lays a solid foundation for future studies aimed at leveraging the beneficial properties of S. epidermidis while mitigating its pathogenic potential.
Collapse
Affiliation(s)
- Nantia Leonidou
- Institute for Bioinformatics and Medical Informatics (IBMI), Eberhard Karl University of Tübingen, Tübingen, Germany
- Department of Computer Science, Eberhard Karl University of Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), Tübingen, Germany
- Quantitative Biology Center (QBiC), Eberhard Karl University of Tübingen, Tübingen, Germany
- Division Systems Biology of Signal Transduction, German Cancer Research Center (DKFZ), Heidelberg, Baden-Württemberg, Germany
| | - Alina Renz
- Institute for Bioinformatics and Medical Informatics (IBMI), Eberhard Karl University of Tübingen, Tübingen, Germany
- Department of Computer Science, Eberhard Karl University of Tübingen, Tübingen, Germany
| | - Benjamin Winnerling
- Institute for Pharmaceutical Microbiology, University of Bonn, Bonn, North Rhine-Westphalia, Germany
- German Center for Infection Research (DZIF), Bonn, Germany
| | - Anastasiia Grekova
- Structural and Computational Biology Unit, European Molecular Biology Laboratory (EMBL), Heidelberg, Baden-Württemberg, Germany
| | - Fabian Grein
- Institute for Pharmaceutical Microbiology, University of Bonn, Bonn, North Rhine-Westphalia, Germany
- German Center for Infection Research (DZIF), Bonn, Germany
| | - Andreas Dräger
- Institute for Bioinformatics and Medical Informatics (IBMI), Eberhard Karl University of Tübingen, Tübingen, Germany
- German Center for Infection Research (DZIF), Tübingen, Germany
- Quantitative Biology Center (QBiC), Eberhard Karl University of Tübingen, Tübingen, Germany
- Data Analytics and Bioinformatics, Institute of Computer Science, Martin Luther University Halle-Wittenberg, Halle (Saale), Germany
| |
Collapse
|
8
|
Sawicki R, Zabost A, Jankowski G, Augustynowicz-Kopeć E, Truszkiewicz W, Sieniawska E. Usnic acid impacts energy production and iron metabolism in Mycobacterium tuberculosis H37Rv. mSystems 2025; 10:e0025625. [PMID: 40202336 PMCID: PMC12090724 DOI: 10.1128/msystems.00256-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/01/2025] [Accepted: 03/05/2025] [Indexed: 04/10/2025] Open
Abstract
Mycobacterium tuberculosis has developed a wide array of response mechanisms to various stress factors. Usnic acid has been demonstrated to be a potent antimycobacterial agent that induces stress responses and growth inhibition in many mycobacterial species. Previous studies have shown that it alters the expression of stress-responsive sigma factors, as well as the metabolites and lipid profile in M. tuberculosis H37Ra. This study was designed to examine potential differences in the strain-specific susceptibility of the virulent H37Rv strain to usnic acid. By combining lipidomic and transcriptomic analyses, we uncovered the impact of usnic acid on bacterial metabolism. The observed downregulation of key lipid classes suggested reduced metabolic activity. The simultaneous elevation of mycobactins-siderophores used by members of the genus Mycobacterium to transport free extracellular iron ions into the cytoplasm-indicated the involvement of iron in the stress response generated by usnic acid. The repressed tricarboxylic acid (TCA) cycle and oxidative phosphorylation were compensated by the upregulation of alternative energy production pathways, such as cytochrome P450 and the ferredoxin reductase system. This indicates that mycobacteria may switch to alternative electron transport mechanisms under usnic acid stress using iron-sulfur clusters to generate energy. From a therapeutic perspective, the study highlights iron metabolism as an essential drug target in mycobacteria. Simultaneously, the results confirm the strain-specific metabolic response of sister strains against the same stressing agent. IMPORTANCE A previous study on the influence of usnic acid on the avirulent H37Ra strain revealed that the early bacterial response was associated with redox homeostasis, lipid synthesis, and nucleic acid repair. The response of bacteria to antimicrobials is specific to each species and strain. Given the genetic and phenotypic differences between the avirulent H37Ra strain and the virulent H37Rv strain, we combined lipidomics and global transcriptomics to uncover the mechanism of action of usnic acid against H37Rv. The study identified strain-specific differences between the virulent H37Rv and avirulent H37Ra. The H37Ra strain exhibited increased metabolic activity, while the H37Rv strain showed a reduction in basic metabolic processes and activated alternative iron-dependent energy production. These differences highlight the varying susceptibility of sister strains within the same species to the same antibacterial agent.
Collapse
Affiliation(s)
- Rafał Sawicki
- Department of Biochemistry and Biotechnology, Medical University of Lublin, Lublin, Poland
| | - Anna Zabost
- Department of Microbiology, National Tuberculosis and Lung Diseases Research Institute, Warsaw, Poland
| | - Grzegorz Jankowski
- Department of Biochemistry and Biotechnology, Medical University of Lublin, Lublin, Poland
| | - Ewa Augustynowicz-Kopeć
- Department of Microbiology, National Tuberculosis and Lung Diseases Research Institute, Warsaw, Poland
| | - Wiesław Truszkiewicz
- Department of Biochemistry and Biotechnology, Medical University of Lublin, Lublin, Poland
| | - Elwira Sieniawska
- Department of Natural Products Chemistry, Medical University of Lublin, Lublin, Poland
| |
Collapse
|
9
|
Parra I, Carrasco-Carballo A, Palafox-Sanchez V, Martínez-García I, Aguilera J, Góngora-Alfaro JL, Aranda-González II, Tizabi Y, Mendieta L. Peroxisome Proliferator-Activated Receptors (PPARs) May Mediate the Neuroactive Effects of Probiotic Metabolites: An In Silico Approach. Int J Mol Sci 2025; 26:4507. [PMID: 40429654 PMCID: PMC12111801 DOI: 10.3390/ijms26104507] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/15/2025] [Revised: 02/25/2025] [Accepted: 03/04/2025] [Indexed: 05/29/2025] Open
Abstract
It is well established that the gut-brain axis (GBA) is a bidirectional communication between the gut and the brain. This axis, critical in maintaining overall homeostasis, is regulated at the neuronal, endocrine, and immunological levels, all of which may be influenced by the gut microbiota (GM). Therefore, dysbiosis or disruption in the GM may have serious consequences including neuroinflammation due to overactivation of the immune system. Strategies to reestablish GM integrity via use of probiotics are being pursued as novel therapeutic intervention in a variety of central and peripheral diseases. The mechanisms leading to dysbiosis or efficacy of probiotics, however, are not fully evident. Here, we performed computational analysis on two major probiotics, namely Lactobacillus Lacticaseibacillus rhamnosus GG (formerly named Lactobacillus rhamnosus, L. rhamnosus GG) and Bifidobacterium animalis spp. lactis (B. lactis or B. animalis) to not only shed some light on their mechanism(s) of action but also to identify potential molecular targets for novel probiotics. Using the PubMed web page and BioCyc Database Collection platform we specifically analyzed proteins affected by metabolites of these bacteria. Our results indicate that peroxisome proliferator-activated receptors (PPARs), nuclear receptor proteins that are involved in regulation of inflammation are key mediators of the neuroactive effect of probiotics.
Collapse
Affiliation(s)
- Irving Parra
- Laboratorio de Neuroquímica, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla 72592, Mexico; (I.P.); (I.M.-G.)
| | - Alan Carrasco-Carballo
- Laboratorio de Elucidación y Síntesis en Química Orgánica, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla 72592, Mexico;
| | - Victoria Palafox-Sanchez
- Institute for Obesity Research, Instituto Tecnológico y de Estudios Superiores de Monterrey, Monterrey 64700, Mexico;
| | - Isabel Martínez-García
- Laboratorio de Neuroquímica, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla 72592, Mexico; (I.P.); (I.M.-G.)
| | - José Aguilera
- Institut de Neurociències, Universitat Autònoma de Barcelona, 08193 Barcelona, Spain;
| | - José L. Góngora-Alfaro
- Departamento de Neurociencias, Centro de Investigaciones Regionales “Dr. Hideyo Noguchi”, Universidad Autónoma de Yucatán, Avenida Itzáes No. 490 x 59, Mérida 97000, Mexico;
| | - Irma Isela Aranda-González
- Facultad de Medicina, Universidad Autónoma de Yucatán, Avenida Itzáes No. 498 x 59 y 59A, Mérida 97000, Mexico;
| | - Yousef Tizabi
- Department of Pharmacology, Howard University College of Medicine, Washington, DC 20059, USA;
| | - Liliana Mendieta
- Laboratorio de Neuroquímica, Facultad de Ciencias Químicas, Benemérita Universidad Autónoma de Puebla, Puebla 72592, Mexico; (I.P.); (I.M.-G.)
| |
Collapse
|
10
|
Gilet L, Leroy M, Maes A, Condon C, Braun F. Unconventional mRNA processing and degradation pathways for the polycistronic yrzI (spyTA) mRNA in Bacillus subtilis. FEBS Lett 2025; 599:1222-1235. [PMID: 40055937 PMCID: PMC12067863 DOI: 10.1002/1873-3468.70027] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/19/2024] [Revised: 02/07/2025] [Accepted: 02/10/2025] [Indexed: 05/13/2025]
Abstract
The ribosome-associated endoribonuclease Rae1 cleaves the Bacillus subtilis yrzI operon mRNA in a translation-dependent manner. This operon encodes up to four small peptides, S1027, YrzI, S1025, and S1024, whose functions are unknown. Here, we identified the function of YrzI and S1025 and deciphered the degradation pathways of the yrzI polycistronic mRNA. We show that YrzI is toxic at high concentrations, but co-expression with S1025 abolishes its toxicity, and that, in the absence of Rae1, S1025 is the major antidote to the YzI toxin. We show that a highly stable mRNA species containing the YrzI and S1025 open reading frames results from endoribonucleolytic cleavage upstream of yrzI followed by the arrest of 5'-exoribonucleolytic processing by ribosomes bound to its exceptionally strong Shine-Dalgarno sequence. Degradation of this mRNA requires either translation-dependent cleavage within S1025 by Rae1 or direct attack from the structured 3'-end by 3'-exoribonucleases. Neither pathway is common for a B. subtilis mRNA.
Collapse
Affiliation(s)
- Laetitia Gilet
- Expression Génétique Microbienne, CNRS ‐ Université Paris Cité, Institut de Biologie Physico‐ChimiqueParisFrance
| | - Magali Leroy
- Expression Génétique Microbienne, CNRS ‐ Université Paris Cité, Institut de Biologie Physico‐ChimiqueParisFrance
| | - Alexandre Maes
- Plateforme de Génomique Fonctionnel, UMR8226 CNRS Sorbonne Université, Institut de Biologie Physico‐ChimiqueParisFrance
| | - Ciarán Condon
- Expression Génétique Microbienne, CNRS ‐ Université Paris Cité, Institut de Biologie Physico‐ChimiqueParisFrance
| | - Frédérique Braun
- Expression Génétique Microbienne, CNRS ‐ Université Paris Cité, Institut de Biologie Physico‐ChimiqueParisFrance
| |
Collapse
|
11
|
Ruhl IA, Woodworth SP, Haugen SJ, Alt HM, Beckham GT, Johnson CW. Production of Vanillin From Ferulic Acid by Pseudomonas putida KT2440 Using Metabolic Engineering and In Situ Product Recovery. Microb Biotechnol 2025; 18:e70152. [PMID: 40410945 PMCID: PMC12102074 DOI: 10.1111/1751-7915.70152] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/11/2024] [Revised: 04/01/2025] [Accepted: 04/16/2025] [Indexed: 05/26/2025] Open
Abstract
Vanillin is the most in-demand flavouring compound in the world and because vanillin extracted from vanilla pods cannot meet the global demand, most vanillin on the market today is chemically synthesised. Increasing demands by consumers for natural ingredients have inspired efforts to develop vanillin derived from microbial sources. These efforts have been challenged by low titers, likely caused by the toxicity of vanillin to most microbial biocatalysts. In this study, we engineered a Pseudomonas putida KT2440-derived strain that accumulated vanillin from ferulic acid to 0.64 g/L. To increase the overall titre, we applied a hydrophobic polystyrene-based resin to vanillin-accumulating cultures, which enabled an increase in total vanillin recovery to an apparent titre of 3.35 g/L. This study demonstrates that P. putida can accumulate vanillin from ferulic acid to higher titers when vanillin is removed from the cultivation medium, mitigating its toxicity.
Collapse
Affiliation(s)
- Ilona A. Ruhl
- Renewable Resources and Enabling Sciences CenterNational Renewable Energy LaboratoryGoldenColoradoUSA
| | - Sean P. Woodworth
- Renewable Resources and Enabling Sciences CenterNational Renewable Energy LaboratoryGoldenColoradoUSA
| | - Stefan J. Haugen
- Renewable Resources and Enabling Sciences CenterNational Renewable Energy LaboratoryGoldenColoradoUSA
| | - Hannah M. Alt
- Renewable Resources and Enabling Sciences CenterNational Renewable Energy LaboratoryGoldenColoradoUSA
| | - Gregg T. Beckham
- Renewable Resources and Enabling Sciences CenterNational Renewable Energy LaboratoryGoldenColoradoUSA
| | - Christopher W. Johnson
- Renewable Resources and Enabling Sciences CenterNational Renewable Energy LaboratoryGoldenColoradoUSA
| |
Collapse
|
12
|
Van Loon JC, Le Mauff F, Vargas MA, Gilbert S, Pfoh R, Morrison ZA, Razvi E, Nitz M, Sheppard DC, Howell PL. Structural and functional analysis of Pseudomonas aeruginosa PelA provides insight into the modification of the Pel exopolysaccharide. J Biol Chem 2025; 301:108432. [PMID: 40120681 PMCID: PMC12022489 DOI: 10.1016/j.jbc.2025.108432] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/06/2025] [Revised: 03/14/2025] [Accepted: 03/18/2025] [Indexed: 03/25/2025] Open
Abstract
A major biofilm matrix determinant of Pseudomonas aeruginosa is the partially deacetylated α-1,4 linked N-acetylgalactosamine polymer, Pel. After synthesis and transport of the GalNAc polysaccharide across the inner membrane, PelA partially deacetylates and hydrolyzes Pel before its export out of the cell via PelB. While the Pel modification and export proteins are known to interact in the periplasm, it is unclear how the interaction of PelA and PelB coordinates these processes. To determine how PelA modifies the polymer, we determined its structure to 2.1 Å and found a unique arrangement of four distinct domains. We have shown previously that the hydrolase domain exhibits endo-α-1,4-N-acetylgalactosaminidase activity. Characterization of the deacetylase domain revealed that PelA is the founding member of a new carbohydrate esterase family, CE21. Further, we found that the PelAB interaction enhances the deacetylation of N-acetylgalactosamine oligosaccharides. Using the PelA structure in conjunction with AlphaFold2 modeling of the PelAB complex, we propose a model wherein PelB guides Pel to the deacetylase domain of PelA and subsequently to the porin domain of PelB for export. Perturbation or loss of the PelAB interaction would result in less efficient deacetylation and potentially increased Pel hydrolysis. In PelA homologs across many phyla, the predicted structure and active sites are conserved, suggesting a common modification mechanism in Gram-negative bacterial species containing a functional pel operon.
Collapse
Affiliation(s)
- Jaime C Van Loon
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - François Le Mauff
- Department of Microbiology and Immunology, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Infectious Disease and Immunity in Global Health, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; McGill Interdisciplinary Initiative in Infection and Immunity, Montreal, Quebec, Canada; GlycoNET Integrated Services, Microbial Glycomic Node, Montreal, Quebec, Canada
| | - Mario A Vargas
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Stephanie Gilbert
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Roland Pfoh
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada
| | - Zachary A Morrison
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Erum Razvi
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada
| | - Mark Nitz
- Department of Chemistry, University of Toronto, Toronto, Ontario, Canada
| | - Donald C Sheppard
- Department of Microbiology and Immunology, Faculty of Medicine, McGill University, Montreal, Quebec, Canada; Infectious Disease and Immunity in Global Health, Research Institute of the McGill University Health Centre, Montreal, Quebec, Canada; McGill Interdisciplinary Initiative in Infection and Immunity, Montreal, Quebec, Canada
| | - P Lynne Howell
- Program in Molecular Medicine, The Hospital for Sick Children, Toronto, Ontario, Canada; Department of Biochemistry, University of Toronto, Toronto, Ontario, Canada.
| |
Collapse
|
13
|
Mukherjee SD, Batagello C, Adler A, Agudelo J, Zampini A, Suryavanshi M, Nguyen A, Orr T, Dearing D, Monga M, Miller AW. Complex system modeling reveals oxalate homeostasis is driven by diverse oxalate-degrading bacteria. eLife 2025; 14:RP104121. [PMID: 40310467 PMCID: PMC12045624 DOI: 10.7554/elife.104121] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/02/2025] Open
Abstract
Decades of research have made clear that host-associated microbiomes touch all facets of health. However, effective therapies that target the microbiome have been elusive given its inherent complexity. Here, we experimentally examined diet-microbe-host interactions through a complex systems framework, centered on dietary oxalate. Using multiple, independent molecular, rodent, and in vitro experimental models, we found that microbiome composition influenced multiple oxalate-microbe-host interfaces. Importantly, the administration of the oxalate-degrading specialist, Oxalobacter formigenes, was only effective against a poor oxalate-degrading microbiota background and gives critical new insights into why clinical intervention trials with this species exhibit variable outcomes. Data suggest that, while heterogeneity in the microbiome impacts multiple diet-host-microbe interfaces, metabolic redundancy among diverse microorganisms in specific diet-microbe axes is a critical variable that may impact the efficacy of bacteriotherapies, which can help guide patient and probiotic selection criteria in probiotic clinical trials.
Collapse
Affiliation(s)
- Sromona D Mukherjee
- Department of Cardiovascular and Metabolic Sciences, Cleveland ClinicClevelandUnited States
| | - Carlos Batagello
- Division of Urology, Hospital das Clínicas, University of Sao Paulo Medical SchoolSao PauloBrazil
| | - Ava Adler
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland ClinicClevelandUnited States
| | - Jose Agudelo
- Department of Cardiovascular and Metabolic Sciences, Cleveland ClinicClevelandUnited States
| | - Anna Zampini
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland ClinicClevelandUnited States
| | - Mangesh Suryavanshi
- Department of Cardiovascular and Metabolic Sciences, Cleveland ClinicClevelandUnited States
| | - Andrew Nguyen
- M Health Fairview Southdale HospitalEdinaUnited States
| | - Terry Orr
- Department of Biology, New Mexico State UniversityLas CrucesUnited States
| | - Denise Dearing
- School of Biological Sciences, University of UtahSalt Lake CityUnited States
| | - Manoj Monga
- Department of Urology, University of California San DiegoSan DiegoUnited States
| | - Aaron W Miller
- Department of Cardiovascular and Metabolic Sciences, Cleveland ClinicClevelandUnited States
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland ClinicClevelandUnited States
| |
Collapse
|
14
|
Liu M, Blattman SB, Takahashi M, Mandayam N, Jiang W, Oikonomou P, Tavazoie SF, Tavazoie S. Conserved genetic basis for microbial colonization of the gut. Cell 2025; 188:2505-2520.e22. [PMID: 40187346 PMCID: PMC12048274 DOI: 10.1016/j.cell.2025.03.010] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 12/02/2024] [Accepted: 03/06/2025] [Indexed: 04/07/2025]
Abstract
Despite the fundamental importance of gut microbes, the genetic basis of their colonization remains largely unexplored. Here, by applying cross-species genotype-habitat association at the tree-of-life scale, we identify conserved microbial gene modules associated with gut colonization. Across thousands of species, we discovered 79 taxonomically diverse putative colonization factors organized into operonic and non-operonic modules. They include previously characterized colonization pathways such as autoinducer-2 biosynthesis and novel processes including tRNA modification and translation. In vivo functional validation revealed YigZ (IMPACT family) and tRNA hydroxylation protein-P (TrhP) are required for E. coli intestinal colonization. Overexpressing YigZ alone is sufficient to enhance colonization of the poorly colonizing MG1655 E. coli by >100-fold. Moreover, natural allelic variations in YigZ impact inter-strain colonization efficiency. Our findings highlight the power of large-scale comparative genomics in revealing the genetic basis of microbial adaptations. These broadly conserved colonization factors may prove critical for understanding gastrointestinal (GI) dysbiosis and developing therapeutics.
Collapse
Affiliation(s)
- Menghan Liu
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Sydney B Blattman
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Mai Takahashi
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY 10065, USA
| | - Nandan Mandayam
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY 10065, USA
| | - Wenyan Jiang
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Panos Oikonomou
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA
| | - Sohail F Tavazoie
- Laboratory of Systems Cancer Biology, The Rockefeller University, New York, NY 10065, USA
| | - Saeed Tavazoie
- Department of Biological Sciences, Columbia University, New York, NY 10027, USA; Department of Systems Biology, Columbia University, New York, NY 10032, USA; Department of Biochemistry and Molecular Biophysics, Columbia University, New York, NY 10032, USA.
| |
Collapse
|
15
|
Sher D, George EE, Wietz M, Gifford S, Zoccarato L, Weissberg O, Koedooder C, Valiya Kalladi WB, Barreto Filho MM, Mireles R, Malavin S, Liddor Naim M, Idan T, Shrivastava V, Itelson L, Sade D, Abu Hamoud A, Soussan-Farhat Y, Barak N, Karp P, Moore LR. Collaborative metabolic curation of an emerging model marine bacterium, Alteromonas macleodii ATCC 27126. PLoS One 2025; 20:e0321141. [PMID: 40273159 PMCID: PMC12021255 DOI: 10.1371/journal.pone.0321141] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/25/2024] [Accepted: 02/28/2025] [Indexed: 04/26/2025] Open
Abstract
Inferring the metabolic capabilities of an organism from its genome is a challenging process, relying on computationally-derived or manually curated metabolic networks. Manual curation can correct mistakes in the draft network and add missing reactions based on the literature, but requires significant expertise and is often the bottleneck for high-quality metabolic reconstructions. Here, we present a synopsis of a community curation workshop for the model marine bacterium Alteromonas macleodii ATCC 27126 and its genome database in BioCyc, focusing on pathways for utilizing organic carbon and nitrogen sources. Due to the scarcity of biochemical information or gene knock-outs, the curation process relied primarily on published growth phenotypes and bioinformatic analyses, including comparisons with related Alteromonas strains. We report full pathways for the utilization of the algal polysaccharides alginate and pectin in contrast to inconclusive evidence for one-carbon metabolism and mixed acid fermentation, in accordance with the lack of growth on methanol and formate. Pathways for amino acid degradation are ubiquitous across Alteromonas macleodii strains, yet enzymes in the pathways for the degradation of threonine, tryptophan and tyrosine were not identified. Nucleotide degradation pathways are also partial in ATCC 27126. We postulate that demonstrated growth on nitrate as sole nitrogen source proceeds via a nitrate reductase pathway that is a hybrid of known pathways. Our evidence highlights the value of joint and interactive curation efforts, but also shows major knowledge gaps regarding Alteromonas metabolism. The manually-curated metabolic reconstruction is available as a "Tier-2" database on BioCyc.
Collapse
Affiliation(s)
- Daniel Sher
- Department of Marine Biology, Leon H. Charney School of Marine Sciences, University of Haifa, Israel
| | - Emma E. George
- Integrative Oceanography Division, Scripps Institution of Oceanography, University of California, San Diego, La Jolla, California, United States of America
| | - Matthias Wietz
- Alfred Wegener Institute Helmholtz Centre for Polar and Marine Research, Bremerhaven, Germany
- Max Planck Institute for Marine Microbiology, Bremen, Germany
| | - Scott Gifford
- Department of Earth, Marine and Environmental Sciences, The University of North Carolina at Chapel Hill, Chapel Hill, North Carolina, United States of America
| | - Luca Zoccarato
- Institute of Computational Biology, University of Natural Resources and Life Sciences, Vienna, Austria
- Core Facility Bioinformatics, University of Natural Resources and Life Sciences, Vienna, Austria
| | - Osnat Weissberg
- Department of Marine Biology, Leon H. Charney School of Marine Sciences, University of Haifa, Israel
| | - Coco Koedooder
- The Fredy and Nadine Herrmann Institute of Earth Sciences, Hebrew University of Jerusalem, Jerusalem, Israel
- The Interuniversity Institute for Marine Sciences in Eilat, Eilat, Israel
- Israel Oceanographic and Limnological Research, Haifa, Israel
| | | | | | - Raul Mireles
- Department of Plant Pathology and Microbiology, Robert H. Smith Faculty of Agriculture, Food and Environment, The Hebrew University of Jerusalem, Rehovot, Israel,
| | - Stas Malavin
- Israel Oceanographic and Limnological Research, Haifa, Israel
- Zuckerberg Institute for Water Research, Ben-Gurion University of the Negev, Beer-Sheba, Israel
| | - Michal Liddor Naim
- Avram and Stella Goldstein-Goren Department of Biotechnology Engineering, Ben-Gurion University of the Negev, Beer-Sheva, Israel
| | - Tal Idan
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Vibhaw Shrivastava
- Department of Marine Biology, Leon H. Charney School of Marine Sciences, University of Haifa, Israel
| | - Lynne Itelson
- School of Zoology, Faculty of Life Sciences, Tel-Aviv University, Tel-Aviv, Israel
| | - Dagan Sade
- Department of Biomolecular Sciences, The Weizmann Institute of Science, Rehovot, Israel
| | - Alhan Abu Hamoud
- Department of Marine Biology, Leon H. Charney School of Marine Sciences, University of Haifa, Israel
| | - Yara Soussan-Farhat
- Department of Marine Biology, Leon H. Charney School of Marine Sciences, University of Haifa, Israel
| | - Noga Barak
- Department of Marine Biology, Leon H. Charney School of Marine Sciences, University of Haifa, Israel
| | - Peter Karp
- Bioinformatics Research Group, SRI International, Menlo Park, California, United States of America.
| | - Lisa R. Moore
- Bioinformatics Research Group, SRI International, Menlo Park, California, United States of America.
| |
Collapse
|
16
|
Baa-Puyoulet P, Gerlin L, Parisot N, Peignier S, Renoz F, Calevro F, Charles H. ArtSymbioCyc, a metabolic network database collection dedicated to arthropod symbioses: a case study, the tripartite cooperation in Sipha maydis. mSystems 2025; 10:e0014025. [PMID: 40116496 PMCID: PMC12013274 DOI: 10.1128/msystems.00140-25] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/29/2025] [Accepted: 02/20/2025] [Indexed: 03/23/2025] Open
Abstract
Most arthropods live in close association with bacteria. The genomes of associated partners have co-evolved, creating situations of interdependence that are complex to decipher despite the availability of their complete sequences. We developed ArtSymbioCyc, a metabolism-oriented database collection gathering genomic resources for arthropods and their associated bacteria. ArtSymbioCyc uses the powerful tools of the BioCyc community to produce high-quality annotations and to analyze and compare metabolic networks on a genome-wide scale. We used ArtSymbioCyc to study the case of the tripartite symbiosis of the cereal aphid Sipha maydis focusing on amino acid and vitamin metabolisms, as these compounds are known to be important in this strictly phloemophagous insect. We showed that the metabolic pathways of the insect host and its two obligate bacterial associates are interdependent and specialized in the exploitation of Poaceae phloem, particularly for the biosynthesis of sulfur-containing amino acids and most vitamins. This demonstrates that ArtSymbioCyc does not only reveal the individual metabolic capacities of each partner and their respective contributions to the holobiont they constitute but also allows to predict the essential inputs that must come from host nutrition.IMPORTANCEThe evolution has driven the emergence of complex arthropod-microbe symbiotic systems, whose metabolic integration is difficult to unravel. With its user-friendly interface, ArtSymbioCyc (https://artsymbiocyc.cycadsys.org) eases and speeds up the analysis of metabolic networks by enabling precise inference of compound exchanges between associated partners and helps unveil the adaptive potential of arthropods in contexts such as conservation or agricultural control.
Collapse
Affiliation(s)
| | - Léo Gerlin
- INRAE, INSA Lyon, BF2I, UMR203, Villeurbanne, France
| | | | | | - François Renoz
- Biodiversity Research Centre, Earth and Life Institute, UCLouvain, Louvain-la-Neuve, Belgium
| | | | | |
Collapse
|
17
|
Sharma OP. PathwayMind: An innovative tool and database for pathway perturbation analysis, uncovering critical pathways for drugs and target protein sets. Comput Biol Chem 2025; 118:108466. [PMID: 40250332 DOI: 10.1016/j.compbiolchem.2025.108466] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/28/2024] [Revised: 03/28/2025] [Accepted: 04/04/2025] [Indexed: 04/20/2025]
Abstract
Pathway enrichment analysis is a valuable tool for researchers aiming to understand the mechanisms underlying any specific drug or disease associated gene lists derived from biological assays or large-scale genome (omics) experiments. By employing this method, researchers can pinpoint biological pathways that exhibit a higher level of enrichment for a given set of genes than would be anticipated by random chance. These biological pathways play a crucial role in understanding the disease pathophysiology, therapeutic strategy, polypharmacological effects, synergistic mechanisms, and target engagement. However many of the available tools do not fulfill this requirement as most of the available tools consider a flat hierarchy of protein involvement in the pathway and do not consider topological information or importance of molecules in the pathway. Here we propose a novel method to enrich the molecular pathways and prioritize them based on their importance and crucial role in the biological function using the graph and evidence-based approach and customized datasets called PathwayMind. It includes 2648 pathways, 4539 biological events, 2465847 protein-protein interactions and 124717 gene-to-pathway relationships and the role of 3510 unique initial genes in 11,992 molecular pathways. The current manuscript comprises three major steps: The first step is about the data extraction and datasets creation for pathway enrichment, and the second steps comprises pathway perturbation analysis to identify most perturbed biological pathways and the third steps includes validation of this approach along with standalone tools and visualization algorithms which disclose the molecular involvement and improve the interpretability of the results. The end-to-end pathway analysis can be performed in a few minutes to provide complete insights of your target or drug of interest. The current PathwayMind tool and its datasets could be very useful for the molecular scientists and system biologists who are interested to understand the therapeutic effects of their drugs or understanding the involvement of biological pathways for specific gene sets which does not require any prior bioinformatics training.
Collapse
|
18
|
Chemla Y, Levin I, Fan Y, Johnson AA, Coley CW, Voigt CA. Hyperspectral reporters for long-distance and wide-area detection of gene expression in living bacteria. Nat Biotechnol 2025:10.1038/s41587-025-02622-y. [PMID: 40216953 DOI: 10.1038/s41587-025-02622-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/09/2024] [Accepted: 02/27/2025] [Indexed: 04/27/2025]
Abstract
Genetically encoded reporters are suitable for short-distance imaging in the laboratory but not for scanning wide outdoor areas from a distance. Here we introduce hyperspectral reporters (HSRs) designed for hyperspectral imaging cameras that are commonly mounted on unmanned aerial vehicles and satellites. HSR genes encode enzymes that produce a molecule with a unique absorption signature that can be reliably distinguished in hyperspectral images. Quantum mechanical simulations of 20,170 metabolites identified candidate HSRs, leading to the selection of biliverdin IXα and bacteriochlorophyll a for their distinct absorption spectra and biosynthetic feasibility. These genes were integrated into chemical sensor circuits in soil (Pseudomonas putida) and aquatic (Rubrivivax gelatinosus) bacteria. The bacteria were detectable outdoors under ambient light from up to 90 m in a single 4,000-m2 hyperspectral image taken using fixed and unmanned aerial vehicle-mounted cameras. The dose-response functions of the chemical sensors were measured remotely. HSRs enable large-scale studies and applications in ecology, agriculture, environmental monitoring, forensics and defense.
Collapse
Affiliation(s)
- Yonatan Chemla
- Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Itai Levin
- Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Yueyang Fan
- Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Anna A Johnson
- Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Connor W Coley
- Department of Chemical Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA
- Department of Electrical Engineering and Computer Science, Massachusetts Institute of Technology, Cambridge, MA, USA
| | - Christopher A Voigt
- Synthetic Biology Center, Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA, USA.
| |
Collapse
|
19
|
Khan E, Rückert-Reed C, Dahiya G, Tietze L, Fages-Lartaud M, Busche T, Kalinowski J, Shingler V, Lale R. High-resolution mapping of sigma factor DNA-binding sequences using artificial promoters, RNA aptamers, and deep sequencing. Nucleic Acids Res 2025; 53:gkaf306. [PMID: 40239990 PMCID: PMC11997794 DOI: 10.1093/nar/gkaf306] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2024] [Revised: 03/07/2025] [Accepted: 04/03/2025] [Indexed: 04/18/2025] Open
Abstract
The variable sigma (σ) subunit of the bacterial RNA polymerase holoenzyme determines promoter specificity and facilitates open complex formation during transcription initiation. Understanding σ-factor binding sequences is therefore crucial for deciphering bacterial gene regulation. Here, we present a data-driven high-throughput approach that utilizes an extensive library of 1.54 million DNA templates providing artificial promoters and 5' untranslated region sequences for σ-factor DNA-binding motif discovery. This method combines the generation of extensive DNA libraries, in vitro transcription, RNA aptamer, and deep DNA and RNA sequencing. It allows direct assessment of promoter activity, identification of transcription start sites, and quantification of promoter strength based on mRNA production levels. We applied this approach to map σ54 DNA-binding sequences in Pseudomonas putida. Deep sequencing of the enriched RNA pool revealed 64 966 distinct σ54 binding motifs, significantly expanding the known repertoire. This data-driven approach surpasses traditional methods by directly evaluating promoter function and avoiding selection bias based solely on binding affinity. This comprehensive dataset enhances our understanding of σ-factor binding sequences and their regulatory roles, opening avenues for new research in biology and biotechnology.
Collapse
Affiliation(s)
- Essa Ahsan Khan
- Department of Biotechnology and Food Science, Faculty of Natural Sciences, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Christian Rückert-Reed
- Bielefeld University, Center for Biotechnology (CeBiTec), Technology Platform Genomics, Bielefeld 33615, Germany
- Bielefeld University, Medical School OWL, Bielefeld 33615, Germany
| | | | - Lisa Tietze
- Department of Biotechnology and Food Science, Faculty of Natural Sciences, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Maxime Fages-Lartaud
- Department of Biotechnology and Food Science, Faculty of Natural Sciences, Norwegian University of Science and Technology, Trondheim 7491, Norway
| | - Tobias Busche
- Bielefeld University, Center for Biotechnology (CeBiTec), Technology Platform Genomics, Bielefeld 33615, Germany
- Bielefeld University, Medical School OWL, Bielefeld 33615, Germany
| | - Jörn Kalinowski
- Bielefeld University, Center for Biotechnology (CeBiTec), Technology Platform Genomics, Bielefeld 33615, Germany
| | - Victoria Shingler
- Department of Molecular Biology, Umeå University, Umeå 90187, Sweden
| | - Rahmi Lale
- Department of Biotechnology and Food Science, Faculty of Natural Sciences, Norwegian University of Science and Technology, Trondheim 7491, Norway
- Syngens AS, Trondheim 7089, Norway
| |
Collapse
|
20
|
de Crécy-Lagard V, Barahoglu Z, Yuan Y, Boël G, Babor J, Bacusmo JM, Dedon PC, Ho P, Hummels KR, Kearns D. Are Bacterial Processes Dependent on Global Ribosome Pausing Affected by tRNA Modification Defects? J Mol Biol 2025:169107. [PMID: 40210524 DOI: 10.1016/j.jmb.2025.169107] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/14/2024] [Revised: 03/23/2025] [Accepted: 03/24/2025] [Indexed: 04/12/2025]
Abstract
By integrating a literature review with transcriptomic, proteomic, and phenotypic data from two model bacteria, Escherichia coli and Vibrio cholerae, we put forward the hypothesis that defects in tRNA modification broadly impact processes that are evolutionarily tuned to be sensitive to translation speed. These include the translation of regulatory proteins associated with motility, iron homeostasis, and leader peptide-driven attenuation mechanisms. Some of these translation speed-dependent processes are influenced by the absence of a single modification, while others are affected by the absence of multiple modifications. Although further experiments are needed to clarify the mechanisms involved in each case, this work provides a foundational framework to guide future research.
Collapse
Affiliation(s)
- Valérie de Crécy-Lagard
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA; Genetics Institute, University of Florida, Gainesville, FL 32611, USA.
| | - Zeynep Barahoglu
- Institut Pasteur, Université Paris Cité, Epitranscriptomic and Translational Responses to Anti-bacterial Stress, 75015 Paris, France; Expression Génétique Microbienne, CNRS, Université Paris Cité, Institut de Biologie Physico-Chimique, F-75005 Paris, France.
| | - Yifeng Yuan
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA.
| | - Grégory Boël
- Expression Génétique Microbienne, CNRS, Université Paris Cité, Institut de Biologie Physico-Chimique, F-75005 Paris, France.
| | - Jill Babor
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA.
| | - Jo Marie Bacusmo
- Department of Microbiology and Cell Science, University of Florida, Gainesville, FL 32611, USA.
| | - Peter C Dedon
- Singapore-MIT Alliance for Research and Technology, Antimicrobial Resistance IRG, Singapore 138602 Singapore; Department of Biological Engineering, Massachusetts Institute of Technology, Cambridge, MA 02139, USA.
| | - Peiying Ho
- Singapore-MIT Alliance for Research and Technology, Antimicrobial Resistance IRG, Singapore 138602 Singapore
| | | | - Daniel Kearns
- Department of Biology, Indiana University, Bloomington, IN 47405, USA.
| |
Collapse
|
21
|
Bai C, Wu L, Li R, Cao Y, He S, Bo X. Machine Learning-Enabled Drug-Induced Toxicity Prediction. ADVANCED SCIENCE (WEINHEIM, BADEN-WURTTEMBERG, GERMANY) 2025; 12:e2413405. [PMID: 39899688 PMCID: PMC12021114 DOI: 10.1002/advs.202413405] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 10/22/2024] [Revised: 12/25/2024] [Indexed: 02/05/2025]
Abstract
Unexpected toxicity has become a significant obstacle to drug candidate development, accounting for 30% of drug discovery failures. Traditional toxicity assessment through animal testing is costly and time-consuming. Big data and artificial intelligence (AI), especially machine learning (ML), are robustly contributing to innovation and progress in toxicology research. However, the optimal AI model for different types of toxicity usually varies, making it essential to conduct comparative analyses of AI methods across toxicity domains. The diverse data sources also pose challenges for researchers focusing on specific toxicity studies. In this review, 10 categories of drug-induced toxicity is examined, summarizing the characteristics and applicable ML models, including both predictive and interpretable algorithms, striking a balance between breadth and depth. Key databases and tools used in toxicity prediction are also highlighted, including toxicology, chemical, multi-omics, and benchmark databases, organized by their focus and function to clarify their roles in drug-induced toxicity prediction. Finally, strategies to turn challenges into opportunities are analyzed and discussed. This review may provide researchers with a valuable reference for understanding and utilizing the available resources to bridge prediction and mechanistic insights, and further advance the application of ML in drugs-induced toxicity prediction.
Collapse
Affiliation(s)
- Changsen Bai
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- Department of Advanced & Interdisciplinary BiotechnologyAcademy of Military Medical SciencesBeijing100850China
- Tianjin Medical University Cancer Institute and HospitalTianjin300060China
| | - Lianlian Wu
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- Department of Advanced & Interdisciplinary BiotechnologyAcademy of Military Medical SciencesBeijing100850China
| | - Ruijiang Li
- Department of Advanced & Interdisciplinary BiotechnologyAcademy of Military Medical SciencesBeijing100850China
| | - Yang Cao
- Department of Environmental MedicineAcademy of Military Medical SciencesTianjin300050China
| | - Song He
- Department of Advanced & Interdisciplinary BiotechnologyAcademy of Military Medical SciencesBeijing100850China
| | - Xiaochen Bo
- Academy of Medical Engineering and Translational MedicineTianjin UniversityTianjin300072China
- Department of Advanced & Interdisciplinary BiotechnologyAcademy of Military Medical SciencesBeijing100850China
| |
Collapse
|
22
|
Delgado-Nungaray JA, Figueroa-Yáñez LJ, Reynaga-Delgado E, García-Ramírez MA, Aguilar-Corona KE, Gonzalez-Reynoso O. Influence of Amino Acids on Quorum Sensing-Related Pathways in Pseudomonas aeruginosa PAO1: Insights from the GEM iJD1249. Metabolites 2025; 15:236. [PMID: 40278365 PMCID: PMC12029727 DOI: 10.3390/metabo15040236] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/07/2025] [Revised: 03/20/2025] [Accepted: 03/26/2025] [Indexed: 04/26/2025] Open
Abstract
BACKGROUND/OBJECTIVES Amino acids (AAs) play a critical role in diseases such as cystic fibrosis where Pseudomonas aeruginosa PAO1 adapts its metabolism in response to host-derived nutrients. The adaptation influences virulence and complicates antibiotic treatment mainly for the antimicrobial resistance context. D- and L-AAs have been analyzed for their impact on quorum sensing (QS), a mechanism that regulates virulence factors. This research aimed to reconstruct the genome-scale metabolic model (GEM) of P. aeruginosa PAO1 to investigate the metabolic roles of D- and L-AAs in QS-related pathways. METHODS The updated GEM, iJD1249, was reconstructed by using protocols to integrate data from previous models and refined with well-standardized in silico media (LB, M9, and SCFM) to improve flux balance analysis accuracy. The model was used to explore the metabolic impact of D-Met, D-Ala, D-Glu, D-Ser, L-His, L-Glu, L-Arg, and L-Ornithine (L-Orn) at 5 and 50 mM in QS-related pathways, focusing on the effects on bacterial growth and carbon flux distributions. RESULTS Among the tested AAs, D-Met was the only one that did not enhance the growth rate of P. aeruginosa PAO1, while L-Arg and L-Orn increased fluxes in the L-methionine biosynthesis pathway, influencing the metH gene. These findings suggest a differential metabolic role for D-and L-AAs in QS-related pathways. CONCLUSIONS Our results shed some light on the metabolic impact of AAs on QS-related pathways and their potential role in P. aeruginosa virulence. Future studies should assess D-Met as a potential adjuvant in antimicrobial strategies, optimizing the concentration in combination with antibiotics to maximize its therapeutic effectiveness.
Collapse
Affiliation(s)
- Javier Alejandro Delgado-Nungaray
- Chemical Engineering Department, University Center for Exact and Engineering Sciences, University of Guadalajara, Blvd. M. García Barragán # 1451, Guadalajara 44430, Mexico;
| | - Luis Joel Figueroa-Yáñez
- Industrial Biotechnology Unit, Center for Research and Assistance in Technology and Design of the State of Jalisco, A.C. (CIATEJ), Zapopan 45019, Mexico;
| | - Eire Reynaga-Delgado
- Pharmacobiology Department, University Center for Exact and Engineering Sciences, University of Guadalajara, Blvd. M. García Barragán # 1451, Guadalajara 44430, Mexico;
| | - Mario Alberto García-Ramírez
- Electronics Department, University Center for Exact and Engineering Sciences, University of Guadalajara, Blvd. M. García Barragán # 1451, Guadalajara 44430, Mexico;
| | - Karla Esperanza Aguilar-Corona
- Food Engineering and Biotechnology, University Center for Exact and Engineering Sciences, University of Guadalajara, Blvd. M. García Barragán # 1451, Guadalajara 44430, Mexico;
| | - Orfil Gonzalez-Reynoso
- Chemical Engineering Department, University Center for Exact and Engineering Sciences, University of Guadalajara, Blvd. M. García Barragán # 1451, Guadalajara 44430, Mexico;
| |
Collapse
|
23
|
Johnson Z, Anderson D, Cheung MS, Bohutskyi P. Gene network centrality analysis identifies key regulators coordinating day-night metabolic transitions in Synechococcus elongatus PCC 7942 despite limited accuracy in predicting direct regulator-gene interactions. Front Microbiol 2025; 16:1569559. [PMID: 40207147 PMCID: PMC11979508 DOI: 10.3389/fmicb.2025.1569559] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2025] [Accepted: 03/07/2025] [Indexed: 04/11/2025] Open
Abstract
Synechococcus elongatus PCC 7942 is a model organism for studying circadian regulation and bioproduction, where precise temporal control of metabolism significantly impacts photosynthetic efficiency and CO2-to-bioproduct conversion. Despite extensive research on core clock components, our understanding of the broader regulatory network orchestrating genome-wide metabolic transitions remains incomplete. We address this gap by applying machine learning tools and network analysis to investigate the transcriptional architecture governing circadian-controlled gene expression. While our approach showed moderate accuracy in predicting individual transcription factor-gene interactions - a common challenge with real expression data - network-level topological analysis successfully revealed the organizational principles of circadian regulation. Our analysis identified distinct regulatory modules coordinating day-night metabolic transitions, with photosynthesis and carbon/nitrogen metabolism controlled by day-phase regulators, while nighttime modules orchestrate glycogen mobilization and redox metabolism. Through network centrality analysis, we identified potentially significant but previously understudied transcriptional regulators: HimA as a putative DNA architecture regulator, and TetR and SrrB as potential coordinators of nighttime metabolism, working alongside established global regulators RpaA and RpaB. This work demonstrates how network-level analysis can extract biologically meaningful insights despite limitations in predicting direct regulatory interactions. The regulatory principles uncovered here advance our understanding of how cyanobacteria coordinate complex metabolic transitions and may inform metabolic engineering strategies for enhanced photosynthetic bioproduction from CO2.
Collapse
Affiliation(s)
- Zachary Johnson
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, United States
- Department of Biological Systems Engineering, Washington State University, Pullman, WA, United States
| | - David Anderson
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, United States
| | - Margaret S. Cheung
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, United States
- Department of Physics, University of Washington, Seattle, WA, United States
| | - Pavlo Bohutskyi
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, United States
- Department of Biological Systems Engineering, Washington State University, Pullman, WA, United States
| |
Collapse
|
24
|
Jönsson M, Sigrist R, Gren T, Semenov Petrov M, Marcussen NEJ, Svetlova A, Charusanti P, Gockel P, Palsson BO, Yang L, Özdemir E. Machine learning uncovers the transcriptional regulatory network for the production host Streptomyces albidoflavus. Cell Rep 2025; 44:115392. [PMID: 40057950 DOI: 10.1016/j.celrep.2025.115392] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/09/2024] [Revised: 01/16/2025] [Accepted: 02/12/2025] [Indexed: 03/29/2025] Open
Abstract
Streptomyces albidoflavus is a widely used strain for natural product discovery and production through heterologous biosynthetic gene clusters (BGCs). However, the transcriptional regulatory network (TRN) and its impact on secondary metabolism remain poorly understood. Here, we characterize the TRN using independent component analysis on 218 RNA sequencing (RNA-seq) transcriptomes across 88 unique growth conditions. We identify 78 independently modulated sets of genes (iModulons) that quantitatively describe the TRN across diverse conditions. Our analyses reveal (1) TRN adaptation to different growth conditions, (2) conserved and unique characteristics of the TRN across diverse lineages, (3) transcriptional activation of several endogenous BGCs, including surugamide, minimycin, and paulomycin, and (4) inferred functions of 40% of uncharacterized genes in the S. albidoflavus genome. These findings provide a comprehensive and quantitative understanding of the S. albidoflavus TRN, offering a knowledge base for further exploration and experimental validation.
Collapse
Affiliation(s)
- Mathias Jönsson
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet, Building 220, 2800 Kgs. Lyngby, Denmark
| | - Renata Sigrist
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet, Building 220, 2800 Kgs. Lyngby, Denmark
| | - Tetiana Gren
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet, Building 220, 2800 Kgs. Lyngby, Denmark
| | - Mykhaylo Semenov Petrov
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet, Building 220, 2800 Kgs. Lyngby, Denmark
| | - Nils Emil Junge Marcussen
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet, Building 220, 2800 Kgs. Lyngby, Denmark
| | - Anna Svetlova
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet, Building 220, 2800 Kgs. Lyngby, Denmark
| | - Pep Charusanti
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet, Building 220, 2800 Kgs. Lyngby, Denmark
| | - Peter Gockel
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet, Building 220, 2800 Kgs. Lyngby, Denmark
| | - Bernhard O Palsson
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet, Building 220, 2800 Kgs. Lyngby, Denmark; Department of Bioengineering, University of California, San Diego, La Jolla, CA 92093, USA
| | - Lei Yang
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet, Building 220, 2800 Kgs. Lyngby, Denmark.
| | - Emre Özdemir
- Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kemitorvet, Building 220, 2800 Kgs. Lyngby, Denmark.
| |
Collapse
|
25
|
Huang A, Ma J, Zhu H, Qi Y, Jin Y, Zhang M, Yin L, Luo M, Chen S, Xie C, Huang H. Blood metabolites mediate causal inference studies on the effect of gut microbiota on the risk of vascular calcification. J Adv Res 2025:S2090-1232(25)00198-5. [PMID: 40139524 DOI: 10.1016/j.jare.2025.03.038] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/17/2025] [Revised: 03/18/2025] [Accepted: 03/20/2025] [Indexed: 03/29/2025] Open
Abstract
BACKGROUND Emerging evidence indicates a notable connection between gut microbiota and Vascular Calcification (VC). Gut microbiota influences various disease processes through host metabolic pathways; however, the causative link between gut microbiota and VC, along with the potential mediating role of metabolites, is still not well understood. METHODS We leveraged data from the largest Genome-Wide Association Studies (GWAS) concerning gut microbiota, blood metabolites, and VC. To explore the causal relationships among these variables, we conducted two-sample bidirectional Mendelian Randomization (MR) analyses. Furthermore, mediation analyses were conducted to determine if metabolites act as an intermediary in the impact of gut microbiota on VC. In addition, we recruited CKD patients for mass spectrometry and CT examination, and performed a correlation analysis between the expression of blood metabolites and VC score. Finally, we experimentally validated the effects of intermediate metabolites on VC. RESULTS We identified 19 positive gut microbiota species and 52 positive blood metabolites with causal effects on VC. Additionally, the onset of VC was found to induce changes in the abundance of 24 gut microbiota species and 56 metabolites. Further analyses revealed that up to 13 positive gut microbiota species regulate the expression of 20 positive metabolites. Mediation analysis suggests that the gut microbiota g_KLE1615 promotes VC by downregulating the methionine-to-phosphate ratio. Mass spectrometry results indicate that over half of the metabolites identified through MR analysis show altered expression during CKD progression. Among them, 7 metabolites were significantly associated with the progression of VC. Further in vitro experiments confirmed the inhibitory effect of the intermediate metabolite methionine on VC. CONCLUSION Gut microbiota and blood metabolites are causally linked to VC. These findings provide a theoretical basis for microbiome- and metabolome-based therapeutic strategies for targeting VC and enhances our comprehension of the gut-vascular axis.
Collapse
Affiliation(s)
- Aoran Huang
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China
| | - Jianshuai Ma
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China
| | - Huijin Zhu
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China
| | - Yanli Qi
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China
| | - Yang Jin
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China
| | - Mingxuan Zhang
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China
| | - Li Yin
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China
| | - Minhong Luo
- Department of Nephrology, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China
| | - Sifan Chen
- Medical Research Center, Sun Yat-Sen Memorial Hospital, Sun Yat-Sen University, Guangzhou 510000, China
| | - Chen Xie
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China.
| | - Hui Huang
- Department of Cardiology, Joint Laboratory of Guangdong-Hong Kong-Macao Universities for Nutritional Metabolism and Precise Prevention and Control of Major Chronic Diseases, the Eighth Affiliated Hospital of Sun Yat-sen University, Shenzhen 518033, China.
| |
Collapse
|
26
|
Nguyen E, Agbavor C, Steenhaut A, Pratyush MR, Hiller NL, Cahoon LA, Mikheyeva IV, Ng WL, Bridges AA. A small periplasmic protein governs broad physiological adaptations in Vibrio cholerae via regulation of the DbfRS two-component system. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.03.24.645060. [PMID: 40196685 PMCID: PMC11974885 DOI: 10.1101/2025.03.24.645060] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 04/09/2025]
Abstract
Two-component signaling pathways allow bacteria to sense and respond to environmental changes, yet the sensory mechanisms of many remain poorly understood. In the pathogen Vibrio cholerae, the DbfRS two-component system controls the biofilm lifecycle, a critical process for environmental persistence and host colonization. Here, we identified DbfQ, a small periplasmic protein encoded adjacent to dbfRS, as a direct modulator of pathway activity. DbfQ directly binds the sensory domain of the histidine kinase DbfS, shifting it toward phosphatase activity and promoting biofilm dispersal. In contrast, outer membrane perturbations, caused by mutations in lipopolysaccharide biosynthesis genes or membrane-damaging antimicrobials, activate phosphorylation of the response regulator DbfR. Transcriptomic analyses reveal that DbfR phosphorylation leads to broad transcriptional changes spanning genes involved in biofilm formation, central metabolism, peptidoglycan synthesis, and cellular stress responses. Constitutive DbfR phosphorylation imposes severe fitness costs in an infection model, highlighting this pathway as a potential target for anti-infective therapeutics. We find that dbfQRS-like genetic modules are widely present across bacterial phyla, underscoring their broad relevance in bacterial physiology. Collectively, these findings establish DbfQ as a new class of periplasmic regulator that influences two-component signaling and bacterial adaptation.
Collapse
Affiliation(s)
- Emmy Nguyen
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Charles Agbavor
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Anjali Steenhaut
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - M R Pratyush
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - N. Luisa Hiller
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Laty A. Cahoon
- Department of Biological Sciences, University of Pittsburgh, Pittsburgh, PA 15260, USA
| | - Irina V. Mikheyeva
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| | - Wai-Leung Ng
- Department of Molecular Biology and Microbiology, Tufts University School of Medicine, Boston, MA 02111, USA
| | - Andrew A. Bridges
- Department of Biological Sciences, Carnegie Mellon University, Pittsburgh, PA 15213, USA
| |
Collapse
|
27
|
de Jager C, Soliman E, Theus MH. Interrogating mediators of single-cell transcriptional changes in the acute damaged cerebral cortex: Insights into endothelial-astrocyte interactions. Mol Cell Neurosci 2025; 133:104003. [PMID: 40090391 DOI: 10.1016/j.mcn.2025.104003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/26/2024] [Revised: 03/09/2025] [Accepted: 03/11/2025] [Indexed: 03/18/2025] Open
Abstract
Traumatic brain injury (TBI) induces complex cellular and molecular changes, challenging recovery and therapeutic development. Although molecular pathways have been implicated in TBI pathology, the cellular specificity of these mechanisms remains underexplored. Here, we investigate the role of endothelial cell (EC) EphA4, a receptor tyrosine kinase receptor involved in axonal guidance, in modulating cell-specific transcriptomic changes within the damaged cerebral cortex. Utilizing single-cell RNA sequencing (scRNA-seq) in an experimental TBI model, we mapped transcriptional changes across various cell types, with a focus on astrocytes and ECs. Our analysis reveals that EC-specific knockout (KO) of EphA4 triggers significant alterations in astrocyte gene expression and shifts predominate subclusters. We identified six distinct astrocyte clusters (C0-C5) in the damaged cortex including as C0-Mobp/Plp1+; C1-Slc1a3/Clu+; C2-Hbb-bs/Hba-a1/Ndrg2+; C3-GFAP/Lcn2+; C4-Gli3/Mertk+, and C5-Cox8a+. We validate a new Sox9+ cluster expressing Mertk and Gas, which mediates efferocytosis to facilitate apoptotic cell clearance and anti-inflammatory responses. Transcriptomic and CellChat analyses of EC-KO cells highlights upregulation of neuroprotective pathways, including increased amyloid precursor protein (APP) and Gas6. Key pathways predicted to be modulated in astrocytes from EC-KO mice include oxidative phosphorylation and FOXO signaling, mitochondrial dysfunction and ephrin B signaling. Concurrently, metabolic and signaling pathways in endothelial cells-such as ceramide and sphingosine phosphate metabolism and NGF-stimulated transcription-indicate an adaptive response to a metabolically demanding post-injury hypoxic environment. These findings elucidate potential interplay between astrocytic and endothelial responses as well as transcriptional networks underlying cortical tissue damage.
Collapse
Affiliation(s)
- Caroline de Jager
- Translational Biology Medicine and Health Graduate Program, Blacksburg, VA 24061, USA
| | - Eman Soliman
- Department of Biomedical Sciences and Pathobiology, Blacksburg, VA 24061, USA
| | - Michelle H Theus
- Department of Biomedical Sciences and Pathobiology, Blacksburg, VA 24061, USA; Center for Engineered Health, Virginia Tech, Blacksburg, VA 24061, USA.
| |
Collapse
|
28
|
Thapa K, Kinali M, Pei S, Luna A, Babur Ö. Strategies to include prior knowledge in omics analysis with deep neural networks. PATTERNS (NEW YORK, N.Y.) 2025; 6:101203. [PMID: 40182174 PMCID: PMC11963003 DOI: 10.1016/j.patter.2025.101203] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Indexed: 04/05/2025]
Abstract
High-throughput molecular profiling technologies have revolutionized molecular biology research in the past decades. One important use of molecular data is to make predictions of phenotypes and other features of the organisms using machine learning algorithms. Deep learning models have become increasingly popular for this task due to their ability to learn complex non-linear patterns. Applying deep learning to molecular profiles, however, is challenging due to the very high dimensionality of the data and relatively small sample sizes, causing models to overfit. A solution is to incorporate biological prior knowledge to guide the learning algorithm for processing the functionally related input together. This helps regularize the models and improve their generalizability and interpretability. Here, we describe three major strategies proposed to use prior knowledge in deep learning models to make predictions based on molecular profiles. We review the related deep learning architectures, including the major ideas in relatively new graph neural networks.
Collapse
Affiliation(s)
- Kisan Thapa
- Computer Science Department, University of Massachusetts Boston, 100 Morrissey Boulevard, Boston, MA 02125, USA
| | - Meric Kinali
- Computer Science Department, University of Massachusetts Boston, 100 Morrissey Boulevard, Boston, MA 02125, USA
| | - Shichao Pei
- Computer Science Department, University of Massachusetts Boston, 100 Morrissey Boulevard, Boston, MA 02125, USA
| | - Augustin Luna
- Developmental Therapeutics Branch, Center for Cancer Research, National Cancer Institute, NIH, 9000 Rockville Pike, Bathesda, MD 20892, USA
- Computational Biology Branch, National Library of Medicine, NIH, 9000 Rockville Pike, Bathesda, MD 20892, USA
| | - Özgün Babur
- Computer Science Department, University of Massachusetts Boston, 100 Morrissey Boulevard, Boston, MA 02125, USA
| |
Collapse
|
29
|
Isokpehi RD, Simmons SS, Makolo AU, Hollman AL, Adesida SA, Ojo OO, Abioye AO. Insights into Functions of Universal Stress Proteins Encoded by Genomes of Gastric Cancer Pathogen Helicobacter pylori and Related Bacteria. Pathogens 2025; 14:275. [PMID: 40137760 PMCID: PMC11944479 DOI: 10.3390/pathogens14030275] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/05/2025] [Revised: 03/09/2025] [Accepted: 03/11/2025] [Indexed: 03/29/2025] Open
Abstract
The genes that encode the universal stress protein (USP) family domain (pfam00582) aid the survival of bacteria in specific host or habitat-induced stress conditions. Genome sequencing revealed that the genome of Helicobacter pylori, a gastric cancer pathogen, typically contains one USP gene, while related helicobacters have one or two distinct USP genes. However, insights into the functions of Helicobacteraceae (Helicobacter and Wolinella) USP genes are still limited to inferences from large-scale genome sequencing. Thus, we have combined bioinformatics and visual analytics approaches to conduct a more comprehensive data investigation of a set of 1045 universal stress protein sequences encoded in 1014 genomes including 785 Helicobacter pylori genomes. The study generated a representative set of 183 USP sequences consisting of 180 Helicobacter sequences, two Wolinella succinogenes sequences, and a sequence from a related campylobacteria. We used the amino acid residues and positions of the 12 possible functional sites in 1030 sequences to identify 25 functional sites patterns for guiding studies on functional interactions of Helicobacteraceae USPs with ATP and other molecules. Genomic context searches and analysis identified USP genes of gastric and enterohepatic helicobacters that are adjacent or in operons with genes for proteins responsive to DNA-damaging oxidative stress (ATP-dependent proteases: ClpS and ClpA); and DNA uptake proteins (natural competence for transformation proteins: ComB6, ComB7, ComB8, ComB9, ComB10, ComBE, and conjugative transfer signal peptidase TraF). Since transcriptomic evidence indicates that oxidative stress and the presence of virulence-associated genes regulate the transcription of H. pylori USP gene, we recommend further research on Helicobacter USP genes and their neighboring genes in oxidative stress response and virulence of helicobacters. To facilitate the reuse of data and research, we produced interactive analytics resources of a dataset composed of values for variables including phylogeography of H. pylori strains, protein sequence features, and gene neighborhood.
Collapse
Affiliation(s)
- Raphael D. Isokpehi
- Transdisciplinary Data Scholars Development Program, Bethune-Cookman University, Daytona Beach, FL 32114, USA
| | - Shaneka S. Simmons
- Division of Arts and Sciences, Jarvis Christian University, Hawkins, TX 75765, USA
| | - Angela U. Makolo
- University of Ibadan Bioinformatics Group, Department of Computer Science, University of Ibadan, Ibadan 200005, Oyo State, Nigeria
| | | | - Solayide A. Adesida
- Department of Microbiology, Faculty of Science, University of Lagos, Akoka 101017, Lagos State, Nigeria
| | - Olabisi O. Ojo
- Department of Natural Sciences, Albany State University, Albany, GA 31721, USA
| | - Amos O. Abioye
- College of Pharmacy & Health Sciences, Belmont University, Nashville, TN 37212, USA;
| |
Collapse
|
30
|
Zbylicki BR, Cochran S, Weiss DS, Ellermeier CD. Identification of two glycosyltransferases required for synthesis of membrane glycolipids in Clostridioides difficile. mBio 2025; 16:e0351224. [PMID: 39964170 PMCID: PMC11898633 DOI: 10.1128/mbio.03512-24] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/12/2024] [Accepted: 01/21/2025] [Indexed: 02/26/2025] Open
Abstract
Clostridioides difficile infections cause over 12,000 deaths and an estimated one billion dollars in healthcare costs annually in the United States. The cell membrane is an essential structure that is important for protection from the extracellular environment, signal transduction, and transport of nutrients. The polar membrane lipids of C. difficile are ~50% glycolipids, a higher percentage than most other organisms. The glycolipids of C. difficile consist of monohexosyldiradylglycerol (MHDRG) (~14%), dihexosyldiradylglycerol (DHDRG) (~15%), trihexosyldiradylglycerol (THDRG) (~5%), and a unique glycolipid aminohexosyl-hexosyldiradylglycerol (HNHDRG) (~16%). Previously, we found that HexSDF are required for the synthesis of HNHDRG. The enzymes required for the synthesis of MHDRG, DHDRG, and THDRG are not known. In this study, we identified the glycosyltransferases UgtA (CDR20291_0008), which is required for the synthesis of all glycolipids, and UgtB (CDR20291_1186), which is required for the synthesis of DHDRG and THDRG. We propose a model where UgtA synthesizes only MHDRG, HexSDF synthesize HNHDRG from MHDRG, and UgtB synthesizes DHDRG and potentially THDRG from MHDRG. We also report that glycolipids are important for critical cell functions, including sporulation, cell size and morphology, maintaining membrane fluidity, colony morphology, and resistance to some membrane-targeting antimicrobials. IMPORTANCE Clostridioides difficile infections are the leading cause of healthcare-associated diarrhea. C. difficile poses a risk to public health due to its ability to form spores and cause recurrent infections. Glycolipids make up ~50% of the polar lipids in the C. difficile membrane, a higher percentage than other common pathogens and include a unique glycolipid not present in other organisms. Here, we identify glycosyltransferases required for the synthesis of glycolipids in C. difficile and demonstrate the important role glycolipids play in C. difficile physiology.
Collapse
Affiliation(s)
- Brianne R. Zbylicki
- Department of Microbiology and Immunology, Carver College of Medicine University of Iowa, Iowa City, Iowa, USA
| | - Sierra Cochran
- Department of Microbiology and Immunology, Carver College of Medicine University of Iowa, Iowa City, Iowa, USA
| | - David S. Weiss
- Department of Microbiology and Immunology, Carver College of Medicine University of Iowa, Iowa City, Iowa, USA
- Graduate Program in Genetics, University of Iowa, Iowa City, Iowa, USA
| | - Craig D. Ellermeier
- Department of Microbiology and Immunology, Carver College of Medicine University of Iowa, Iowa City, Iowa, USA
- Graduate Program in Genetics, University of Iowa, Iowa City, Iowa, USA
| |
Collapse
|
31
|
Gilliam A, Sadler NC, Li X, Garcia M, Johnson Z, Veličković M, Kim YM, Feng S, Qian WJ, Cheung MS, Bohutskyi P. Cyanobacterial circadian regulation enhances bioproduction under subjective nighttime through rewiring of carbon partitioning dynamics, redox balance orchestration, and cell cycle modulation. Microb Cell Fact 2025; 24:56. [PMID: 40055679 PMCID: PMC11889915 DOI: 10.1186/s12934-025-02665-5] [Citation(s) in RCA: 1] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 01/27/2025] [Indexed: 05/13/2025] Open
Abstract
BACKGROUND The industrial feasibility of photosynthetic bioproduction using cyanobacterial platforms remains challenging due to insufficient yields, particularly due to competition between product formation and cellular carbon demands across different temporal phases of growth. This study investigates how circadian clock regulation impacts carbon partitioning between storage, growth, and product synthesis in Synechococcus elongatus PCC 7942, and provides insights that suggest potential strategies for enhanced bioproduction. RESULTS After entrainment to light-dark cycles, PCC 7942 cultures transitioned to constant light revealed distinct temporal patterns in sucrose production, exhibiting three-fold higher productivity during subjective night compared to subjective day despite moderate down-regulation of genes from the photosynthetic apparatus. This enhanced productivity coincided with reduced glycogen accumulation and halted cell division at subjective night time, suggesting temporal separation of competing processes. Transcriptome analysis revealed coordinated circadian clock-driven adjustment of the cell cycle and rewiring of energy and carbon metabolism, with over 300 genes showing differential expression across four time points. The subjective night was characterized by altered expression of cell division-related genes and reduced expression of genes involved in glycogen synthesis, while showing upregulation of glycogen degradation pathways, alternative electron flow components, the pentose phosphate pathway, and oxidative decarboxylation of pyruvate. These molecular changes created favorable conditions for product formation through enhanced availability of major sucrose precursors (glucose-1-phosphate and fructose-6-phosphate) and maintained redox balance through multiple mechanisms. CONCLUSIONS Our analysis of circadian regulatory rewiring of carbon metabolism and redox balancing suggests two potential approaches that could be developed for improving cyanobacterial bioproduction: leveraging natural circadian rhythms for optimizing cultivation conditions and timing of pathway induction, and engineering strains that mimic circadian-driven metabolic shifts through controlled carbon flux redistribution and redox rebalancing. While these strategies remain to be tested, they could theoretically improve the efficiency of photosynthetic bioproduction by enabling better temporal separation between cell growth, carbon storage accumulation, and product synthesis phases.
Collapse
Affiliation(s)
- Ashley Gilliam
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Natalie C Sadler
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Xiaolu Li
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Marci Garcia
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Zachary Johnson
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
- Department of Biological Systems Engineering, Washington State University, Pullman, WA, USA
| | - Marija Veličković
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Young-Mo Kim
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Song Feng
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Wei-Jun Qian
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA
| | - Margaret S Cheung
- Environmental Molecular Sciences Laboratory, Pacific Northwest National Laboratory, Richland, WA, USA
- Department of Physics, University of Washington, Seattle, WA, USA
| | - Pavlo Bohutskyi
- Biological Sciences Division, Earth and Biological Sciences Directorate, Pacific Northwest National Laboratory, Richland, WA, USA.
- Department of Biological Systems Engineering, Washington State University, Pullman, WA, USA.
| |
Collapse
|
32
|
Oluoch PO, Koh EI, Proulx MK, Reames CJ, Papavinasasundaram KG, Murphy KC, Zimmerman MD, Dartois V, Sassetti CM. Chemical genetic interactions elucidate pathways controlling tuberculosis antibiotic efficacy during infection. Proc Natl Acad Sci U S A 2025; 122:e2417525122. [PMID: 39993187 PMCID: PMC11892619 DOI: 10.1073/pnas.2417525122] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/04/2024] [Accepted: 01/10/2025] [Indexed: 02/26/2025] Open
Abstract
Successful tuberculosis therapy requires treatment with an unwieldy multidrug combination for several months. Thus, there is a growing need to identify novel genetic vulnerabilities that can be leveraged to develop new, more effective antitubercular drugs. Consequently, recent efforts to optimize tuberculosis (TB) therapy have exploited Mycobacterium tuberculosis (Mtb) chemical genetics to identify pathways influencing antibiotic efficacy, novel mechanisms of antibiotic action, and new targets for TB drug discovery. However, the influence of the complex host environment on these interactions remains largely unknown, leaving the therapeutic potential of the identified targets unclear. In this study, we leveraged a library of conditional mutants targeting 467 essential Mtb genes to characterize the chemical-genetic interactions (CGIs) with TB drugs directly in the mouse infection model. We found that these in vivo CGIs differ significantly from those identified in vitro. Both drug-specific and drug-agnostic effects were identified, and many were preserved during treatment with a multidrug combination, suggesting numerous strategies for enhancing therapy. This work also elucidated the complex effects of pyrazinamide (PZA), a drug that relies on aspects of the infection environment for efficacy. Specifically, our work supports the importance of coenzyme A synthesis- inhibition during infection, as well as the antagonistic effect of iron limitation on PZA activity. In addition, we found that inhibition of thiamine and purine synthesis increases PZA efficacy, suggesting additional therapeutically exploitable metabolic dependencies. Our findings present a map of the unique in vivo CGIs, characterizing the mechanism of PZA activity in vivo and identifying potential targets for TB drug development.
Collapse
Affiliation(s)
- Peter O. Oluoch
- Department of Microbiology, University of Massachusetts Medical School, Worcester, MA01655
| | - Eun-Ik Koh
- Department of Microbiology, University of Massachusetts Medical School, Worcester, MA01655
| | - Megan K. Proulx
- Department of Microbiology, University of Massachusetts Medical School, Worcester, MA01655
| | - Charlotte J. Reames
- Department of Microbiology, University of Massachusetts Medical School, Worcester, MA01655
| | | | - Kenan C. Murphy
- Department of Microbiology, University of Massachusetts Medical School, Worcester, MA01655
| | - Matthew D. Zimmerman
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ07110
| | - Véronique Dartois
- Center for Discovery and Innovation, Hackensack Meridian Health, Nutley, NJ07110
| | | |
Collapse
|
33
|
Duan C, Zang Z, Xu Y, He H, Li S, Liu Z, Lei Z, Zheng JS, Li SZ. FGeneBERT: function-driven pre-trained gene language model for metagenomics. Brief Bioinform 2025; 26:bbaf149. [PMID: 40211978 PMCID: PMC11986344 DOI: 10.1093/bib/bbaf149] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/25/2024] [Revised: 02/22/2025] [Accepted: 03/14/2025] [Indexed: 04/14/2025] Open
Abstract
Metagenomic data, comprising mixed multi-species genomes, are prevalent in diverse environments like oceans and soils, significantly impacting human health and ecological functions. However, current research relies on K-mer, which limits the capture of structurally and functionally relevant gene contexts. Moreover, these approaches struggle with encoding biologically meaningful genes and fail to address the one-to-many and many-to-one relationships inherent in metagenomic data. To overcome these challenges, we introduce FGeneBERT, a novel metagenomic pre-trained model that employs a protein-based gene representation as a context-aware and structure-relevant tokenizer. FGeneBERT incorporates masked gene modeling to enhance the understanding of inter-gene contextual relationships and triplet enhanced metagenomic contrastive learning to elucidate gene sequence-function relationships. Pre-trained on over 100 million metagenomic sequences, FGeneBERT demonstrates superior performance on metagenomic datasets at four levels, spanning gene, functional, bacterial, and environmental levels and ranging from 1 to 213 k input sequences. Case studies of ATP synthase and gene operons highlight FGeneBERT's capability for functional recognition and its biological relevance in metagenomic research.
Collapse
Affiliation(s)
- Chenrui Duan
- College of Computer Science and Technology, Zhejiang University, No. 866, Yuhangtang Road, 310058 Zhejiang, P. R. China
- School of Engineering, Westlake University, No. 600 Dunyu Road, 310030 Zhejiang, P. R. China
| | - Zelin Zang
- Centre for Artificial Intelligence and Robotics (CAIR), HKISI-CAS Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong 310000, China
| | - Yongjie Xu
- College of Computer Science and Technology, Zhejiang University, No. 866, Yuhangtang Road, 310058 Zhejiang, P. R. China
- School of Engineering, Westlake University, No. 600 Dunyu Road, 310030 Zhejiang, P. R. China
| | - Hang He
- School of Medicine and School of Life Sciences, Westlake University, No. 600 Dunyu Road, 310030 Zhejiang, P. R. China
| | - Siyuan Li
- College of Computer Science and Technology, Zhejiang University, No. 866, Yuhangtang Road, 310058 Zhejiang, P. R. China
- School of Engineering, Westlake University, No. 600 Dunyu Road, 310030 Zhejiang, P. R. China
| | - Zihan Liu
- College of Computer Science and Technology, Zhejiang University, No. 866, Yuhangtang Road, 310058 Zhejiang, P. R. China
- School of Engineering, Westlake University, No. 600 Dunyu Road, 310030 Zhejiang, P. R. China
| | - Zhen Lei
- Centre for Artificial Intelligence and Robotics (CAIR), HKISI-CAS Hong Kong Institute of Science & Innovation, Chinese Academy of Sciences, Hong Kong 310000, China
- State Key Laboratory of Multimodal Artificial Intelligence Systems (MAIS), Institute of Automation, Chinese Academy of Sciences (CASIA), Beijing 100190, China
- School of Artificial Intelligence, University of Chinese Academy of Sciences (UCAS), Beijing 100049, China
| | - Ju-Sheng Zheng
- School of Medicine and School of Life Sciences, Westlake University, No. 600 Dunyu Road, 310030 Zhejiang, P. R. China
| | - Stan Z Li
- School of Engineering, Westlake University, No. 600 Dunyu Road, 310030 Zhejiang, P. R. China
| |
Collapse
|
34
|
Heck KL, Yi Y, Thornton D, Zheng J, Calderón AI. A comparative metabolomics analysis of Açaí (Euterpe oleracea Mart.) fruit, food powder, and botanical dietary supplement extracts. PHYTOCHEMICAL ANALYSIS : PCA 2025; 36:394-408. [PMID: 38965051 DOI: 10.1002/pca.3416] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 02/15/2024] [Revised: 06/17/2024] [Accepted: 06/22/2024] [Indexed: 07/06/2024]
Abstract
INTRODUCTION Euterpe oleracea Mart. (açaí) is a botanical of interest to many who seek functional foods that provide antioxidant and anti-inflammatory properties. Cancer patients are increasingly taking botanical dietary supplements containing açaí to complement their conventional therapeutics, which may lead to serious adverse events. Before testing our açaí extracts in vitro for botanical-drug interactions, the goal is to chemically characterize our extracts for compounds whose biological activity in açaí is unknown. OBJECTIVE The objective of this work was to develop a chemical fingerprinting method for untargeted characterization of açaí samples from a variety of sources, including food products and botanical dietary supplement capsules, made with multiple extraction solvents. METHODS An optimized LC-MS method was generated for in-depth untargeted fingerprinting of chemical constituents in açaí extracts. Statistical analysis models were used to describe relationships between the açaí extracts based on molecular features found in both positive and negative mode ESI. RESULTS In an attempt to elucidate the differences in metabolites among açaí extracts from different cultivars, we identified or tentatively identified 173 metabolites from the 16 extracts made from 6 different sources. Of these compounds, there are 138 reported in açaí for the first time. Statistical models showed similar yet distinct differences between the extracts tested based on the polarity of compounds present and the origin of the source material. CONCLUSION A high-resolution mass spectrometry method was generated that allowed us to greatly characterize 16 complex extracts made from different sources of açaí with different extraction solvent polarities.
Collapse
Affiliation(s)
- Kabre L Heck
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL, USA
| | - Yuyan Yi
- Department of Mathematics and Statistics, College of Science and Mathematics, Auburn University, AL, USA
| | - Destini Thornton
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL, USA
| | - Jingyi Zheng
- Department of Mathematics and Statistics, College of Science and Mathematics, Auburn University, AL, USA
| | - Angela I Calderón
- Department of Drug Discovery and Development, Harrison College of Pharmacy, Auburn University, Auburn, AL, USA
| |
Collapse
|
35
|
Federici F, Luppino F, Aguilar-Vilar C, Mazaraki ME, Petersen LB, Ahonen L, Nikel PI. CIFR (Clone-Integrate-Flip-out-Repeat): A toolset for iterative genome and pathway engineering of Gram-negative bacteria. Metab Eng 2025; 88:180-195. [PMID: 39778677 DOI: 10.1016/j.ymben.2025.01.001] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2024] [Revised: 01/01/2025] [Accepted: 01/05/2025] [Indexed: 01/11/2025]
Abstract
Advanced genome engineering enables precise and customizable modifications of bacterial species, and toolsets that exhibit broad-host compatibility are particularly valued owing to their portability. Tn5 transposon vectors have been widely used to establish random integrations of desired DNA sequences into bacterial genomes. However, the iteration of the procedure remains challenging because of the limited availability and reusability of selection markers. We addressed this challenge with CIFR, a mini-Tn5 integration system tailored for iterative genome engineering. The pCIFR vectors incorporate attP and attB sites flanking an antibiotic resistance marker used to select for the insertion. Subsequent removal of antibiotic determinants is facilitated by the Bxb1 integrase paired to a user-friendly counter-selection marker, both encoded in auxiliary plasmids. CIFR delivers engineered strains harboring stable DNA insertions and free of any antibiotic resistance cassette, allowing for the reusability of the tool. The system was validated in Pseudomonas putida, Escherichia coli, and Cupriavidus necator, underscoring its portability across diverse industrially relevant hosts. The CIFR toolbox was calibrated through combinatorial integrations of chromoprotein genes in P. putida, generating strains displaying a diverse color palette. We also introduced a carotenoid biosynthesis pathway in P. putida in a two-step engineering process, showcasing the potential of the tool for pathway balancing. The broad utility of the CIFR toolbox expands the toolkit for metabolic engineering, allowing for the construction of complex phenotypes while opening new possibilities in bacterial genetic manipulations.
Collapse
Affiliation(s)
- Filippo Federici
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Francesco Luppino
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Clara Aguilar-Vilar
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Maria Eleni Mazaraki
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Lars Boje Petersen
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Linda Ahonen
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark
| | - Pablo I Nikel
- The Novo Nordisk Foundation Center for Biosustainability, Technical University of Denmark, Kongens Lyngby, Denmark.
| |
Collapse
|
36
|
Zhang Z, Guo Q, Yang Z, Sun Y, Jiang S, He Y, Li J, Zhang J. Bifidobacterium adolescentis-derived nicotinic acid improves host skeletal muscle mitochondrial function to ameliorate sarcopenia. Cell Rep 2025; 44:115265. [PMID: 39908139 DOI: 10.1016/j.celrep.2025.115265] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/16/2024] [Revised: 11/29/2024] [Accepted: 01/13/2025] [Indexed: 02/07/2025] Open
Abstract
Sarcopenia significantly diminishes quality of life and increases mortality risk in older adults. While the connection between the gut microbiome and muscle health is recognized, the underlying mechanisms are poorly understood. In this study, shotgun metagenomics revealed that Bifidobacterium adolescentis is notably depleted in individuals with sarcopenia, correlating with reduced muscle mass and function. This finding was validated in aged mice. Metabolomics analysis identified nicotinic acid as a key metabolite produced by B. adolescentis, linked to improvements in muscle mass and functionality in individuals with sarcopenia. Mechanistically, nicotinic acid restores nicotinamide adenine dinucleotide (NAD+) levels in muscle, inhibits the FoxO3/Atrogin-1/Murf-1 axis, and promotes satellite cell proliferation, reducing muscle atrophy. Additionally, NAD+ activation enhances the silent-information-regulator 1 (SIRT1)/peroxisome-proliferator-activated-receptor-γ-coactivator 1-alpha (PGC-1α) axis, stimulating mitochondrial biogenesis and promoting oxidative metabolism in slow-twitch fibers, ultimately improving muscle function. Our findings suggest that B. adolescentis-derived nicotinic acid could be a promising therapeutic strategy for individuals with sarcopenia.
Collapse
Affiliation(s)
- Zeng Zhang
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou 570228, Hainan, China
| | - Quan Guo
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou 570228, Hainan, China
| | - Zhihan Yang
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou 570228, Hainan, China
| | - Yukai Sun
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou 570228, Hainan, China
| | - Shuaiming Jiang
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou 570228, Hainan, China
| | - Yangli He
- Department of Health Center, Hainan General Hospital, Hainan Affiliated Hospital of Hainan Medical University, Haikou 570311, Hainan, China
| | - Jiahe Li
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou 570228, Hainan, China
| | - Jiachao Zhang
- School of Food Science and Engineering, Key Laboratory of Food Nutrition and Functional Food of Hainan Province, Hainan University, Haikou 570228, Hainan, China; Collaborative Innovation Center of One Health, Hainan University, Haikou 570228, Hainan, China.
| |
Collapse
|
37
|
Mukherjee SD, Batagello CA, Adler A, Agudelo J, Zampini A, Suryavanshi M, Nguyen A, Orr T, Dearing D, Monga M, Miller AW. Complex system modelling reveals oxalate homeostasis is driven by diverse oxalate-degrading bacteria. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.10.28.620613. [PMID: 39553961 PMCID: PMC11565779 DOI: 10.1101/2024.10.28.620613] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2024]
Abstract
Decades of research have made clear that host-associated microbiomes touch all facets of health. However, effective therapies that target the microbiome have been elusive given its inherent complexity. Here, we experimentally examined diet-microbe-host interactions through a complex systems framework, centered on dietary oxalate. Using multiple, independent molecular, animal, and in vitro experimental models, we found that microbiome composition influenced multiple oxalate-microbe-host interfaces. Importantly, administration of the oxalate-degrading specialist, Oxalobacter formigenes, was only effective against a poor oxalate-degrading microbiota background and gives critical new insights into why clinical intervention trials with this species exhibit variable outcomes. Data suggest that, while heterogeneity in the microbiome impacts multiple diet-host-microbe interfaces, metabolic redundancy among diverse microorganisms in specific diet-microbe axes is a critical variable that may impact the efficacy of bacteriotherapies, which can help guide patient and probiotic selection criteria in probiotic clinical trials.
Collapse
Affiliation(s)
- Sromona D. Mukherjee
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - Carlos A. Batagello
- Division of Urology, Hospital das Clínicas, University of Sao Paulo Medical School, Sao Paulo, Brazil
| | - Ava Adler
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Jose Agudelo
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - Anna Zampini
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
| | - Mangesh Suryavanshi
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, OH, USA
| | - Andrew Nguyen
- M Health Fairview Southdale Hospital, Edina, MN, USA
| | - Teri Orr
- Department of Biology, New Mexico State University, Las Cruces, NM, USA
| | - Denise Dearing
- School of Biological Sciences, University of Utah, Salt Lake City, UT, USA
| | - Manoj Monga
- Department of Urology, University of California San Diego, San Diego, CA, USA
| | - Aaron W. Miller
- Department of Cardiovascular and Metabolic Sciences, Cleveland Clinic, Cleveland, OH, USA
- Department of Urology, Glickman Urological and Kidney Institute, Cleveland Clinic, Cleveland, OH, USA
| |
Collapse
|
38
|
Koper P, Wysokiński J, Żebracki K, Decewicz P, Dziewit Ł, Kalita M, Palusińska-Szysz M, Mazur A. Comparative analysis of Legionella lytica genome identifies specific metabolic traits and virulence factors. Sci Rep 2025; 15:5554. [PMID: 39952999 PMCID: PMC11828895 DOI: 10.1038/s41598-025-90154-5] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/29/2024] [Accepted: 02/11/2025] [Indexed: 02/17/2025] Open
Abstract
The complete genome of Legionella lytica PCM 2298 was sequenced and analyzed to provide insights into its genomic structure, virulence potential, and evolutionary position within the Legionella genus. The genome comprised a 3.2 Mbp chromosome and two plasmids, pLlyPCM2298_1 and pLlyPCM2298_2, contributing to a total genome size of 3.7 Mbp. Functional annotation identified 3,165 coding sequences, including genes associated with known virulence factors such as the major outer membrane protein (MOMP), the macrophage infectivity potentiator (Mip), and a comprehensive set of secretion systems (type II, type IVA, and type IVB Dot/Icm type IV secretion system). Notably, L. lytica contributed 383 unique genes to the Legionella pangenome, with 232 identified effector proteins, of which 35 were plasmid-encoded. The identification of unique genes, particularly those on plasmids, suggests an evolutionary strategy favoring horizontal gene transfer and niche adaptation. The effector repertoire included proteins with domains characteristic of host interaction strategies, such as ankyrin repeats and protein kinases. Comparative analyses showed that while L. lytica shares core virulence traits with other Legionella species, it has distinct features that may contribute to its adaptability and pathogenic potential. These findings underscore the genomic diversity within the genus and contribute to a deeper understanding of Legionella's ecological and clinical significance. A custom web application was developed using the R Shiny library, enabling users to interactively explore the expanded Legionella pangenome through UpSet plots.
Collapse
Affiliation(s)
- Piotr Koper
- Department of Genetics and Microbiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Lublin, Poland.
- Bioinformatics and Biostatistics Laboratory, Institute of Biological Sciences, Maria Curie-Skłodowska University, Lublin, Poland.
| | - Jakub Wysokiński
- Department of Genetics and Microbiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Lublin, Poland
| | - Kamil Żebracki
- Department of Genetics and Microbiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Lublin, Poland
| | - Przemysław Decewicz
- Department of Environmental Microbiology and Biotechnology, Institute of Microbiology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Łukasz Dziewit
- Department of Environmental Microbiology and Biotechnology, Institute of Microbiology, Faculty of Biology, University of Warsaw, Warsaw, Poland
| | - Michał Kalita
- Department of Genetics and Microbiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Lublin, Poland
| | - Marta Palusińska-Szysz
- Department of Genetics and Microbiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Lublin, Poland
| | - Andrzej Mazur
- Department of Genetics and Microbiology, Institute of Biological Sciences, Maria Curie-Skłodowska University, Lublin, Poland
| |
Collapse
|
39
|
Zhang S, Liu K, Liu Y, Hu X, Gu X. The role and application of bioinformatics techniques and tools in drug discovery. Front Pharmacol 2025; 16:1547131. [PMID: 40017606 PMCID: PMC11865229 DOI: 10.3389/fphar.2025.1547131] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2024] [Accepted: 01/27/2025] [Indexed: 03/01/2025] Open
Abstract
The process of drug discovery and development is both lengthy and intricate, demanding a substantial investment of time and financial resources. Bioinformatics techniques and tools can not only accelerate the identification of drug targets and the screening and refinement of drug candidates, but also facilitate the characterization of side effects and the prediction of drug resistance. High-throughput data from genomics, transcriptomics, proteomics, and metabolomics make significant contributions to mechanics-based drug discovery and drug reuse. This paper summarizes bioinformatics technologies and tools in drug research and development and their roles and applications in drug research and development, aiming to provide references for the development of new drugs and the realization of precision medicine.
Collapse
Affiliation(s)
- Shujun Zhang
- Department of Infectious Diseases, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Kaijie Liu
- Department of Infectious Diseases, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Yafeng Liu
- Department of Infectious Diseases, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| | - Xinjun Hu
- Department of Infectious Diseases, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, China
- Henan Medical Key Laboratory of Gastrointestinal Microecology and Hepatology, Luoyang, China
| | - Xinyu Gu
- Department of Oncology, The First Affiliated Hospital, College of Clinical Medicine, Henan University of Science and Technology, Luoyang, Henan, China
| |
Collapse
|
40
|
Beale DJ, Nguyen TV, Dyall T, van de Kamp J, Bissett A, Hewitt L, Small AH. Use of fecal microbiome to understand the impact of housing conditions on metabolic stress responses in farmed saltwater crocodiles ( Crocodylus porosus). Front Vet Sci 2025; 12:1496946. [PMID: 40018705 PMCID: PMC11865912 DOI: 10.3389/fvets.2025.1496946] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2024] [Accepted: 01/13/2025] [Indexed: 03/01/2025] Open
Abstract
Introduction Understanding the impact of housing conditions on the stress responses in farmed saltwater crocodiles (Crocodylus porosus) is crucial for optimizing welfare and management practices. Methods This study employed a multi-omics methodology, combining targeted and untargeted LC-MS for metabolite, lipid, and hormone profiling with 16S rRNA gene sequencing for microbiome analysis, to compare stress responses and changes in fecal samples of crocodiles housed in single versus group pens. Metabolic responses to a startle test were evaluated through multivariate analysis, and changes post-stress were examined. Results A total of 564 metabolic features were identified. Of these, 15 metabolites were linked to the cortisol biosynthesis pathway. Metabolite origin analysis showed that 128 metabolites originated from the host, 151 from the microbiota, and 400 remained unmatched. No significant differences in fecal corticosterone levels were observed between single and group pens. However, metabolic profiling revealed distinct differences in stress responses: single pen crocodiles exhibited downregulation of certain compounds and upregulation of others, affecting pyrimidine and purine metabolism pathways when compared to grouped pen crocodiles, linked to altering energy associated induced stress. Additionally, fecal microbiome analysis indicated increased Firmicutes:Bacteroides (F:B) ratio in group-housed animals, suggesting greater stress. Discussion The study highlights that while traditional stress indicators like corticosterone levels may not differ significantly between housing conditions, metabolic and microbiome analyses provide deeper insights into stress responses. Single pens are associated with less metabolic disruption and potentially better health outcomes compared to group pens. These findings underscore the value of fecal microbiome and metabolomics in assessing animal welfare in farmed crocodiles.
Collapse
Affiliation(s)
- David J. Beale
- Environment, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Ecosciences Precinct, Dutton Park, QLD, Australia
| | - Thao V. Nguyen
- Environment, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Ecosciences Precinct, Dutton Park, QLD, Australia
| | - Tim Dyall
- Agriculture & Food, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Armidale, NSW, Australia
| | - Jodie van de Kamp
- Environment, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Battery Point, TAS, Australia
| | - Andrew Bissett
- Environment, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Battery Point, TAS, Australia
| | - Leisha Hewitt
- Roseworthy Campus, School of Animal and Veterinary Sciences, The University of Adelaide, Roseworthy, Australia
| | - Alison H. Small
- Agriculture & Food, Commonwealth Scientific and Industrial Research Organisation (CSIRO), Armidale, NSW, Australia
| |
Collapse
|
41
|
Liu Z, You J, Zhao P, Wang X, Sun S, Wang X, Gu S, Xu Q. Metabolomics Profiling and Advanced Methodologies for Wheat Stress Research. Metabolites 2025; 15:123. [PMID: 39997748 PMCID: PMC11857233 DOI: 10.3390/metabo15020123] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/16/2024] [Revised: 02/05/2025] [Accepted: 02/06/2025] [Indexed: 02/26/2025] Open
Abstract
Metabolomics is an omics technology that studies the types, quantities, and changes of endogenous metabolic substances in organisms affected by abiotic and biotic factors. BACKGROUND/OBJECTIVES Based on metabolomics, small molecule metabolites in biological organisms can be qualitatively and quantitatively analysed. This method analysis directly correlates with biological phenotypes, facilitating the interpretation of life conditions. Wheat (Triticum aestivum L.) is one of the major food crops in the world, and its quality and yield play important roles in safeguarding food security. METHODS This review elaborated on the significance of metabolomics research techniques and methods in enhancing wheat resilience against biotic and abiotic stresses. RESULTS Metabolomics plays an important role in identifying the metabolites in wheat that respond to diverse stresses. The integrated examination of metabolomics with other omics disciplines provides new insights and approaches for exploring resistance genes, understanding the genetic basis of wheat metabolism, and revealing the mechanisms involved in stress responses. CONCLUSIONS Emerging metabolomics research techniques to propose innovative avenues of research is important to enhance wheat resistance.
Collapse
Affiliation(s)
- Zhen Liu
- National Key Laboratory of Wheat Improvement, College of Agronomy, Shandong Agricultural University, Taian 271018, China; (Z.L.); (P.Z.); (S.S.)
| | - Jiahui You
- Shandong Guocangjian Biotechnology Co., Ltd., Taian 271018, China; (J.Y.); (X.W.); (X.W.)
| | - Peiying Zhao
- National Key Laboratory of Wheat Improvement, College of Agronomy, Shandong Agricultural University, Taian 271018, China; (Z.L.); (P.Z.); (S.S.)
| | - Xianlin Wang
- Shandong Guocangjian Biotechnology Co., Ltd., Taian 271018, China; (J.Y.); (X.W.); (X.W.)
| | - Shufang Sun
- National Key Laboratory of Wheat Improvement, College of Agronomy, Shandong Agricultural University, Taian 271018, China; (Z.L.); (P.Z.); (S.S.)
| | - Xizhen Wang
- Shandong Guocangjian Biotechnology Co., Ltd., Taian 271018, China; (J.Y.); (X.W.); (X.W.)
| | - Shubo Gu
- National Key Laboratory of Wheat Improvement, College of Agronomy, Shandong Agricultural University, Taian 271018, China; (Z.L.); (P.Z.); (S.S.)
| | - Qian Xu
- National Key Laboratory of Wheat Improvement, College of Agronomy, Shandong Agricultural University, Taian 271018, China; (Z.L.); (P.Z.); (S.S.)
| |
Collapse
|
42
|
Raustad N, Dai Y, Iinishi A, Mohapatra A, Soo M, Hay E, Hernandez G, Geisinger E. A phosphorylation signal activates genome-wide transcriptional control by BfmR, the global regulator of Acinetobacter resistance and virulence. Nucleic Acids Res 2025; 53:gkaf063. [PMID: 39921563 PMCID: PMC11806355 DOI: 10.1093/nar/gkaf063] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/30/2024] [Revised: 01/17/2025] [Accepted: 01/23/2025] [Indexed: 02/10/2025] Open
Abstract
The nosocomial pathogen Acinetobacter baumannii is a major threat to human health. The sensor kinase-response regulator system, BfmS-BfmR, is essential to multidrug resistance and virulence in the bacterium and represents a potential antimicrobial target. Important questions remain about how the system controls resistance and pathogenesis. Although BfmR knockout alters expression of >1000 genes, its direct regulon is undefined. Moreover, how phosphorylation controls the regulator is unclear. Here, we address these problems by combining mutagenesis, ChIP-seq, and in vitro phosphorylation to study the functions of phospho-BfmR. We show that phosphorylation is required for BfmR-mediated gene regulation, antibiotic resistance, and sepsis development in vivo. Consistent with activating the protein, phosphorylation induces dimerization and target DNA affinity. Integrated analysis of genome-wide binding and transcriptional profiles of BfmR led to additional key findings: (1) Phosphorylation dramatically expands the number of genomic sites BfmR binds; (2) DNA recognition involves a direct repeat motif widespread across promoters; (3) BfmR directly regulates 303 genes as activator (e.g., capsule, peptidoglycan, and outer membrane biogenesis) or repressor (pilus biogenesis); (4) BfmR controls several non-coding sRNAs. These studies reveal the centrality of a phosphorylation signal in driving A. baumannii disease and disentangle the extensive pathogenic gene-regulatory network under its control.
Collapse
Affiliation(s)
- Nicole Raustad
- Department of Biology, Northeastern University, Boston, MA 02115, United States
| | - Yunfei Dai
- Department of Biology, Northeastern University, Boston, MA 02115, United States
| | - Akira Iinishi
- Antimicrobial Discovery Center, Department of Biology, Northeastern University, Boston, MA 02115, United States
| | - Arpita Mohapatra
- Department of Biology, Northeastern University, Boston, MA 02115, United States
| | - Mark W Soo
- Department of Biology, Northeastern University, Boston, MA 02115, United States
| | - Everett Hay
- Department of Biology, Northeastern University, Boston, MA 02115, United States
| | | | - Edward Geisinger
- Department of Biology, Northeastern University, Boston, MA 02115, United States
| |
Collapse
|
43
|
Palmer-Rodríguez P, Alberich R, Reyes-Prieto M, Castro JA, Llabrés M. Metadag: a web tool to generate and analyse metabolic networks. BMC Bioinformatics 2025; 26:31. [PMID: 39875845 PMCID: PMC11776228 DOI: 10.1186/s12859-025-06048-w] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2024] [Accepted: 01/13/2025] [Indexed: 01/30/2025] Open
Abstract
BACKGROUND MetaDAG is a web-based tool developed to address challenges posed by big data from omics technologies, particularly in metabolic network reconstruction and analysis. The tool is capable of constructing metabolic networks for specific organisms, sets of organisms, reactions, enzymes, or KEGG Orthology (KO) identifiers. By retrieving data from the KEGG database, MetaDAG helps users visualize and analyze complex metabolic interactions efficiently. RESULTS MetaDAG computes two models: a reaction graph and a metabolic directed acyclic graph (m-DAG). The reaction graph represents reactions as nodes and metabolite flow between them as edges. The m-DAG simplifies the reaction graph by collapsing strongly connected components, significantly reducing the number of nodes while maintaining connectivity. MetaDAG can generate metabolic networks from various inputs, including KEGG organisms or custom data (e.g., reactions, enzymes, KOs). The tool displays these models on an interactive web page and provides downloadable files, including network visualizations. MetaDAG was tested using two datasets. In an eukaryotic analysis, it successfully classified organisms from the KEGG database at the kingdom and phylum levels. In a microbiome study, MetaDAG accurately distinguished between Western and Korean diets and categorized individuals by weight loss outcomes based on dietary interventions. CONCLUSION MetaDAG offers an effective and versatile solution for metabolic network reconstruction from diverse data sources, enabling large-scale biological comparisons. Its ability to generate synthetic metabolisms and its broad application, from taxonomy classification to diet analysis, make it a valuable tool for biological research. MetaDAG is available online, with user support provided via a comprehensive guide. MetaDAG: https://bioinfo.uib.es/metadag/ User guide: https://biocom-uib.github.io/MetaDag/.
Collapse
Affiliation(s)
- Pere Palmer-Rodríguez
- Mathematics and Computer Science Department, University of the Balearic Islands, Ctra Valldemossa, Km 7.5, Palma, 07122, Balearic Islands, Spain.
| | - Ricardo Alberich
- Mathematics and Computer Science Department, University of the Balearic Islands, Ctra Valldemossa, Km 7.5, Palma, 07122, Balearic Islands, Spain
| | - Mariana Reyes-Prieto
- Sequencing and Bioinformatics Service, Foundation for the Promotion of Health and Biomedical Research of the Valencian Community (FISABIO), Avda. de Catalunya, 21, 46020, Valencia, Valencia, Spain
| | - José A Castro
- Biology Department, University of the Balearic Islands, Ctra Valldemossa, Km 7.5, 07122, Palma, Balearic Islands, Spain
| | - Mercè Llabrés
- Mathematics and Computer Science Department, University of the Balearic Islands, Ctra Valldemossa, Km 7.5, Palma, 07122, Balearic Islands, Spain.
| |
Collapse
|
44
|
Park J, Jang M, Choi E, Lee SM, Bang I, Woo J, Kim S, Lee EJ, Kim D. ChIP-mini: a low-input ChIP-exo protocol for elucidating DNA-binding protein dynamics in intracellular pathogens. Nucleic Acids Res 2025; 53:gkaf009. [PMID: 39868540 PMCID: PMC11770342 DOI: 10.1093/nar/gkaf009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/25/2024] [Revised: 12/04/2024] [Accepted: 01/06/2025] [Indexed: 01/28/2025] Open
Abstract
Genome-wide identification of binding profiles for DNA-binding proteins from the limited number of intracellular pathogens in infection studies is crucial for understanding virulence and cellular processes but remains challenging, as the current ChIP-exo is designed for high-input bacterial cells (>1010). Here, we developed an optimized ChIP-mini method, a low-input ChIP-exo utilizing a 5,000-fold reduced number of initial bacterial cells and an analysis pipeline, to identify genome-wide binding dynamics of DNA-binding proteins in host-infected pathogens. Applying ChIP-mini to intracellular Salmonella Typhimurium, we identified 642 and 1,837 binding sites of H-NS and RpoD, respectively, elucidating changes in their binding position and binding intensity during infection. Post-infection, we observed 21 significant reductions in H-NS binding at intergenic regions, exposing the promoter region of virulence genes, such as those in Salmonella pathogenicity islands-2, 3 and effectors. Furthermore, we revealed the crucial phenomenon that novel and significantly increased RpoD bindings were found within regions exhibiting diminished H-NS binding, thereby facilitating substantial upregulation of virulence genes. These findings markedly enhance our understanding of how H-NS and RpoD simultaneously coordinate the transcription initiation of virulence genes within macrophages. Collectively, this work demonstrates a broadly adaptable tool that will enable the elucidation of DNA-binding protein dynamics in diverse intracellular pathogens during infection.
Collapse
Affiliation(s)
- Joon Young Park
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Minchang Jang
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Eunna Choi
- Department of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Sang-Mok Lee
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Ina Bang
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Jihoon Woo
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| | - Seonggyu Kim
- Department of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Eun-Jin Lee
- Department of Life Sciences, College of Life Sciences and Biotechnology, Korea University, Seoul, 02841, Republic of Korea
| | - Donghyuk Kim
- School of Energy and Chemical Engineering, Ulsan National Institute of Science and Technology (UNIST), Ulsan, 44919, Republic of Korea
| |
Collapse
|
45
|
Nauta KM, Gates DR, Weiland M, Mechan-Llontop ME, Wang X, Nguyen KP, Isaguirre C, Genjdar MR, Sheldon RD, Krawczyk CM, Burton NO. A noncanonical polyamine from bacteria antagonizes animal mitochondrial function. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2024.04.29.591726. [PMID: 38746390 PMCID: PMC11092615 DOI: 10.1101/2024.04.29.591726] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Indexed: 05/16/2024]
Abstract
Canonical polyamines such as agmatine, putrescine, and spermidine are evolutionarily conserved metabolites found in nearly all forms of life ranging from bacteria to humans. Recently, interactions between polyamines produced by gut bacteria and human intestinal cells have been proposed to contribute to both Irritable Bowel Syndrome with Diarrhea (IBS-D) and inflammatory bowel diseases. However, the molecular mechanisms that underlie these effects are often unclear due in part to limitations in the methods used to manipulate and study polyamine functions in vivo. Here, we developed a Caenorhabditis elegans based screening platform and a modified LC-MS approach for profiling polyamine metabolites. We combined these methods to make the unexpected discovery that dysfunctional polyamine metabolism in both Gram-negative (E. coli) and Gram-positive (B. subtilis) bacteria can result in the accumulation of a noncanonical polyamine intermediate, N1-Aminopropylagmatine (N1-APA). We further find that N1-APA is produced via spermidine synthase (SpeE) and that it is bioactive when encountered by animals. Specifically, we find that when N1-APA is produced by bacteria in animal intestines it can be transported into intestinal cells via the polyamine transporter CATP-5 where it antagonizes both animal development and mitochondrial function across diverse animal species. Lastly, we find that N1-APA functions analogously to the deoxyhypusine synthase inhibitor GC7. For example, like GC7, N1-APA antagonizes eIF5A hypusination and inhibits the alternative activation of mammalian macrophages. To our knowledge, these findings are the first to demonstrate that N1-APA is a bioactive metabolite and that bacteria can produce a small molecule that functions similarly to existing deoxyhypusine synthase inhibitors. Furthermore, these results suggest an exciting new mechanistic hypothesis for why the loss of speB in gut microbes, including E. coli, has been both linked to inflammatory bowel disease (IBD) in humans and found to drive IBD in germ free mice.
Collapse
Affiliation(s)
- Kelsie M. Nauta
- Van Andel Research Institute, Department of Metabolism and Nutritional Programing, Grand Rapids, MI, 49503, USA
| | - Darrick R. Gates
- Van Andel Research Institute, Department of Metabolism and Nutritional Programing, Grand Rapids, MI, 49503, USA
| | - Matthew Weiland
- Van Andel Research Institute, Department of Metabolism and Nutritional Programing, Grand Rapids, MI, 49503, USA
| | - Marco E. Mechan-Llontop
- Van Andel Research Institute, Department of Metabolism and Nutritional Programing, Grand Rapids, MI, 49503, USA
| | - Xiao Wang
- Van Andel Research Institute, Department of Metabolism and Nutritional Programing, Grand Rapids, MI, 49503, USA
| | - Kim P. Nguyen
- Van Andel Research Institute, Department of Metabolism and Nutritional Programing, Grand Rapids, MI, 49503, USA
| | - Christine Isaguirre
- Van Andel Research Institute, Mass Spectrometry Core, Grand Rapids, MI, 49503, USA
| | - Megan R. Genjdar
- Van Andel Research Institute, Mass Spectrometry Core, Grand Rapids, MI, 49503, USA
| | - Ryan D. Sheldon
- Van Andel Research Institute, Mass Spectrometry Core, Grand Rapids, MI, 49503, USA
| | - Connie M. Krawczyk
- Van Andel Research Institute, Department of Metabolism and Nutritional Programing, Grand Rapids, MI, 49503, USA
| | - Nicholas O. Burton
- Van Andel Research Institute, Department of Metabolism and Nutritional Programing, Grand Rapids, MI, 49503, USA
| |
Collapse
|
46
|
Zbylicki BR, Cochran S, Weiss DS, Ellermeier CD. Identification of two glycosyltransferases required for synthesis of membrane glycolipids in Clostridioides difficile. BIORXIV : THE PREPRINT SERVER FOR BIOLOGY 2025:2025.01.14.632984. [PMID: 39868222 PMCID: PMC11761805 DOI: 10.1101/2025.01.14.632984] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Download PDF] [Subscribe] [Scholar Register] [Indexed: 01/28/2025]
Abstract
Clostridioides difficile infections cause over 12,000 deaths and an estimated one billion dollars in healthcare costs annually in the United States. The cell membrane is an essential structure that is important for protection from the extracellular environment, signal transduction, and transport of nutrients. The polar membrane lipids of C. difficile are ~50% glycolipids, a higher percentage than most other organisms. The glycolipids of C. difficile consist of monohexosyldiradylglycerol (MHDRG) (~14%), dihexosyldiradylglycerol (DHDRG) (~15%), trihexosyldiradylglycerol (THDRG) (~5%), and a unique glycolipid aminohexosyl-hexosyldiradylglycerol (HNHDRG) (~16%). Previously, we found HexSDF are required for synthesis of HNHDRG. The enzymes required for synthesis of MHDRG, DHDRG, and THDRG are not known. In this study, we identified the glycosyltransferases UgtA (CDR20291_0008), which is required for synthesis of all glycolipids, and UgtB (CDR20291_1186), which is required for synthesis of DHDRG and THDRG. We propose a model where UgtA synthesizes only MHDRG, HexSDF synthesize HNHDRG from MHDRG, and UgtB synthesizes DHDRG and potentially THDRG from MHDRG. We also report that glycolipids are important for critical cell functions, including sporulation, cell size and morphology, maintaining membrane fluidity, colony morphology, and resistance to some membrane targeting antimicrobials.
Collapse
Affiliation(s)
- Brianne R. Zbylicki
- Department of Microbiology and Immunology Carver College of Medicine University of Iowa 431 Newton Rd Iowa City, IA 52242
| | - Sierra Cochran
- Department of Microbiology and Immunology Carver College of Medicine University of Iowa 431 Newton Rd Iowa City, IA 52242
| | - David S. Weiss
- Department of Microbiology and Immunology Carver College of Medicine University of Iowa 431 Newton Rd Iowa City, IA 52242
- Graduate Program in Genetics, University of Iowa, Iowa City, IA 52242, USA
| | - Craig D. Ellermeier
- Department of Microbiology and Immunology Carver College of Medicine University of Iowa 431 Newton Rd Iowa City, IA 52242
- Graduate Program in Genetics, University of Iowa, Iowa City, IA 52242, USA
| |
Collapse
|
47
|
Johnson S, Weigele P, Fomenkov A, Ge A, Vincze A, Eaglesham J, Roberts R, Sun Z. Domainator, a flexible software suite for domain-based annotation and neighborhood analysis, identifies proteins involved in antiviral systems. Nucleic Acids Res 2025; 53:gkae1175. [PMID: 39657740 PMCID: PMC11754643 DOI: 10.1093/nar/gkae1175] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2024] [Revised: 11/07/2024] [Accepted: 11/15/2024] [Indexed: 12/12/2024] Open
Abstract
The availability of large databases of biological sequences presents an opportunity for in-depth exploration of gene diversity and function. Bacterial defense systems are a rich source of diverse but difficult to annotate genes with biotechnological applications. In this work, we present Domainator, a flexible and modular software suite for domain-based gene neighborhood and protein search, extraction and clustering. We demonstrate the utility of Domainator through three examples related to bacterial defense systems. First, we cluster CRISPR-associated Rossman fold (CARF) containing proteins with difficult to annotate effector domains, classifying most of them as likely transcriptional regulators and a subset as likely RNases. Second, we extract and cluster P4-like phage satellite defense hotspots, identify an abundant variant of Lamassu defense systems and demonstrate its in vivo activity against several T-even phages. Third, we integrate a protein language model into Domainator and use it to identify restriction endonucleases with low similarity to known reference sequences, validating the activity of one example in vitro. Domainator is made available as an open-source package with detailed documentation and usage examples.
Collapse
Affiliation(s)
| | | | | | - Andrew Ge
- New England Biolabs Inc., Ipswich, MA 01938, USA
| | - Anna Vincze
- New England Biolabs Inc., Ipswich, MA 01938, USA
| | | | | | - Zhiyi Sun
- New England Biolabs Inc., Ipswich, MA 01938, USA
| |
Collapse
|
48
|
Müller J, Bayer FP, Wilhelm M, Schuh MG, Kuster B, The M. PTMNavigator: interactive visualization of differentially regulated post-translational modifications in cellular signaling pathways. Nat Commun 2025; 16:510. [PMID: 39779715 PMCID: PMC11711753 DOI: 10.1038/s41467-024-55533-y] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/28/2023] [Accepted: 12/16/2024] [Indexed: 01/11/2025] Open
Abstract
Post-translational modifications (PTMs) play pivotal roles in regulating cellular signaling, fine-tuning protein function, and orchestrating complex biological processes. Despite their importance, the lack of comprehensive tools for studying PTMs from a pathway-centric perspective has limited our ability to understand how PTMs modulate cellular pathways on a molecular level. Here, we present PTMNavigator, a tool integrated into the ProteomicsDB platform that offers an interactive interface for researchers to overlay experimental PTM data with pathway diagrams. PTMNavigator provides ~3000 canonical pathways from manually curated databases, enabling users to modify and create custom diagrams tailored to their data. Additionally, PTMNavigator automatically runs kinase and pathway enrichment algorithms whose results are directly integrated into the visualization. This offers a comprehensive view of the intricate relationship between PTMs and signaling pathways. We demonstrate the utility of PTMNavigator by applying it to two phosphoproteomics datasets, showing how it can enhance pathway enrichment analysis, visualize how drug treatments result in a discernable flow of PTM-driven signaling, and aid in proposing extensions to existing pathways. By enhancing our understanding of cellular signaling dynamics and facilitating the discovery of PTM-pathway interactions, PTMNavigator advances our knowledge of PTM biology and its implications in health and disease.
Collapse
Affiliation(s)
- Julian Müller
- Proteomics and Bioanalytics, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Florian P Bayer
- Proteomics and Bioanalytics, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Mathias Wilhelm
- Computational Mass Spectrometry, School of Life Sciences, Technical University of Munich, Freising, Germany
| | - Maximilian G Schuh
- Proteomics and Bioanalytics, School of Life Sciences, Technical University of Munich, Freising, Germany
- Organic Chemistry II, School of Natural Sciences, Technical University of Munich, Garching, Germany
| | - Bernhard Kuster
- Proteomics and Bioanalytics, School of Life Sciences, Technical University of Munich, Freising, Germany
- German Cancer Consortium (DKTK), Partner Site Munich, Munich, Germany
| | - Matthew The
- Proteomics and Bioanalytics, School of Life Sciences, Technical University of Munich, Freising, Germany.
| |
Collapse
|
49
|
Catoiu EA, Krishnan J, Li G, Lou XA, Rychel K, Yuan Y, Bajpe H, Patel A, Choe D, Shin J, Burrows J, Phaneuf P, Zielinski DC, Palsson BO. iModulonDB 2.0: dynamic tools to facilitate knowledge-mining and user-enabled analyses of curated transcriptomic datasets. Nucleic Acids Res 2025; 53:D99-D106. [PMID: 39494532 PMCID: PMC11701608 DOI: 10.1093/nar/gkae1009] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/26/2024] [Revised: 10/04/2024] [Accepted: 10/22/2024] [Indexed: 11/05/2024] Open
Abstract
iModulons-sets of co-expressed genes identified through independent component analysis (ICA) of high-quality transcriptomic datasets-provide an unbiased, modular view of an organism's transcriptional regulatory network. Established in 2020, iModulonDB (iModulonDB.org) serves as a centralized repository of curated iModulon sets, enabling users to explore iModulons and download the associated transcriptomic data. This update reflects a significant expansion of the database-19 new ICA decompositions (+633%) spanning 8 925 expression profiles (+1370%), 503 studies (+2290%) and 12 additional organisms (+400%)-and introduces new features to help scientists decipher the mechanisms governing prokaryotic transcriptional regulation. To facilitate comprehension of the underlying expression profiles, the updated user-interface displays essential information about each data-generating study (e.g. the experimental conditions and publication abstract). Dashboards now include condition-specific coloring and highlight data generated from genetically perturbed strains, enabling users to rapidly interpret disruptions in transcriptional regulation. New interactive graphs rapidly convey omics-derived indicators (e.g. the explained variance of ICA decompositions, genetic overlap between iModulons and regulons). Direct links to operon diagrams (BioCyc) and protein-protein interaction networks (STRING) provide users with seamless access to external resources for further assessment of iModulons. Lastly, a new suite of search-driven and species-wide analysis tools promotes user-engagement with iModulons, reinforcing iModulonDB's role as a dynamic, interactive knowledgebase of prokaryotic transcriptional regulation.
Collapse
Affiliation(s)
- Edward A Catoiu
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92101, USA
| | - Jayanth Krishnan
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92101, USA
| | - Gaoyuan Li
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92101, USA
| | - Xuwen A Lou
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92101, USA
| | - Kevin Rychel
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92101, USA
| | - Yuan Yuan
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92101, USA
| | - Heera Bajpe
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92101, USA
| | - Arjun Patel
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92101, USA
| | - Donghui Choe
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92101, USA
| | - Jongoh Shin
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92101, USA
| | - Joshua Burrows
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92101, USA
| | - Patrick V Phaneuf
- The Novo Nordisk Foundation (NNF) Center for Biosustainability, The Technical University of Denmark, Kongens Lyngby 2800, Denmark
| | - Daniel C Zielinski
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92101, USA
| | - Bernhard O Palsson
- Department of Bioengineering, University of California, San Diego, La Jolla, CA 92101, USA
- The Novo Nordisk Foundation (NNF) Center for Biosustainability, The Technical University of Denmark, Kongens Lyngby 2800, Denmark
| |
Collapse
|
50
|
Wang X, Yang Q, Shi C, Wang Y, Guo D, Wan X, Dong P, Zhang Q, Hu Y, Zhang R, Yang H, Chen W, Liu Z. Carbon dioxide enhances Akkermansia muciniphila fitness and anti-obesity efficacy in high-fat diet mice. THE ISME JOURNAL 2025; 19:wraf034. [PMID: 39987558 PMCID: PMC11931619 DOI: 10.1093/ismejo/wraf034] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Subscribe] [Scholar Register] [Received: 10/01/2024] [Revised: 12/23/2024] [Accepted: 02/20/2025] [Indexed: 02/25/2025]
Abstract
Numerous studies and clinical applications have underscored the therapeutic potential of the indigenous gut bacterium Akkermansia muciniphila in various diseases. However, our understanding of how Akkermansia muciniphila senses and responds to host gastrointestinal signals remains limited. Here, we demonstrate that A. muciniphila exhibits rapid growth, facilitated by its self-produced carbon dioxide (CO₂), with key enzymes such as glutamate decarboxylase, carbonic anhydrase, and pyruvate ferredoxin oxidoreductase playing pivotal roles. Additionally, we design a novel delivery system, comprising calcium carbonate, inulin, A. muciniphila, and sodium alginate, which enhances A. muciniphila growth and facilitates the expression of part probiotic genes in mice intestinal milieu. Notably, the administration of this delivery system induces weight loss in mice fed high-fat diets. Furthermore, we elucidate the significant impact of CO₂ on the composition and functional genes of the human gut microbiota, with genes encoding carbonic anhydrase and amino acid metabolism enzymes exhibiting heightened responsiveness. These findings reveal a novel mechanism by which gut commensal bacteria sense and respond to gaseous molecules, thereby promoting growth. Moreover, they suggest the potential for designing rational therapeutic strategies utilizing live bacterial delivery systems to enhance probiotic growth and ameliorate gut microbiota-related diseases.
Collapse
Affiliation(s)
- Xiangfeng Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Luoyu Road 1037, Hongshan District, Wuhan, Hubei 430074, China
| | - Qianqian Yang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Luoyu Road 1037, Hongshan District, Wuhan, Hubei 430074, China
| | - Changping Shi
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Luoyu Road 1037, Hongshan District, Wuhan, Hubei 430074, China
| | - Yuyang Wang
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Luoyu Road 1037, Hongshan District, Wuhan, Hubei 430074, China
| | - Dingming Guo
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Luoyu Road 1037, Hongshan District, Wuhan, Hubei 430074, China
| | - Xuchun Wan
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Luoyu Road 1037, Hongshan District, Wuhan, Hubei 430074, China
| | - Pengyuan Dong
- Xi’an Jiaotong-Liverpool University, 111 Ren’ai Road, Suzhou Industrial Park, Suzhou, Jiangsu 215123, China
| | - Qianyao Zhang
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, 1168 West Chunrong Road, Yuhua Avenue, Chenggong District, Kunming Medical University, Kunming 650500, China
| | - Yueyan Hu
- Division of geriatric Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Xichang Road No. 153, Wuhua District, Kunming, Yunnan 650032, China
| | - Ruilin Zhang
- NHC Key Laboratory of Drug Addiction Medicine, School of Forensic Medicine, 1168 West Chunrong Road, Yuhua Avenue, Chenggong District, Kunming Medical University, Kunming 650500, China
| | - Hongju Yang
- Division of geriatric Gastroenterology, The First Affiliated Hospital of Kunming Medical University, Xichang Road No. 153, Wuhua District, Kunming, Yunnan 650032, China
| | - Weihua Chen
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Center for Artificial Intelligence Biology, Department of Bioinformatics and Systems Biology, College of Life Science and Technology, Huazhong University of Science and Technology, Luoyu Road 1037, Hongshan District, Wuhan, Hubei 430074, China
| | - Zhi Liu
- Key Laboratory of Molecular Biophysics of the Ministry of Education, Hubei Key Laboratory of Bioinformatics and Molecular Imaging, Department of Biotechnology, College of Life Science and Technology, Huazhong University of Science and Technology, Luoyu Road 1037, Hongshan District, Wuhan, Hubei 430074, China
| |
Collapse
|